1
|
Cheong DHJ, Yi B, Wong YH, Chu JJH. The Current Progress in the Quest for Vaccines Against the Semliki Forest Virus Complex. Med Res Rev 2025. [PMID: 39757142 DOI: 10.1002/med.22097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 10/10/2024] [Accepted: 12/12/2024] [Indexed: 01/07/2025]
Abstract
The Semliki Forest virus (SFV) complex comprises of arboviruses that are transmitted by arthropod vectors and cause acute febrile illness in humans. In the last seven decades, re-emergence of these viruses has resulted in numerous outbreaks globally, affecting regions including Africa, Americas, Asia, Europe and the Caribbean. These viruses are transmitted to humans by the bite of infected mosquitoes. Symptoms of infection include high fever, severe joint pain, skin rash, muscle pain and headache. Fatal cases were reported, and mortality rate increased during the epidemic of these viruses. There is therefore a need to control the spread of these emerging arboviruses. Given that vaccination is one of the most effective ways to protect populations against viral outbreaks, efforts have been made to develop and test potential vaccine candidates. However, there are still no licensed vaccines available against the medically important viruses in the SFV complex. This review first summarizes the current knowledge of the SFV complex disease pathogenesis. Next, seven strategies that have been applied in vaccine development against these viruses are reviewed, indicating the immune response and efficacies of these vaccine candidates in in vivo models of infection. Finally, the more promising candidates that have entered clinical trials are discussed and insights into the future development of vaccines for viruses of the SFV complex are given.
Collapse
Affiliation(s)
- Dorothy Hui Juan Cheong
- Department of Microbiology and Immunology, Infectious Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Bowen Yi
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Yi Hao Wong
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Justin Jang Hann Chu
- Department of Microbiology and Immunology, Infectious Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
- Collaborative and Translation Unit for Hand, Foot and Mouth Disease (HFMD), Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore CIty, Singapore
| |
Collapse
|
2
|
Freppel W, Silva LA, Stapleford KA, Herrero LJ. Pathogenicity and virulence of chikungunya virus. Virulence 2024; 15:2396484. [PMID: 39193780 PMCID: PMC11370967 DOI: 10.1080/21505594.2024.2396484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted, RNA virus that causes an often-severe musculoskeletal illness characterized by fever, joint pain, and a range of debilitating symptoms. The virus has re-emerged as a global health threat in recent decades, spreading from its origin in Africa across Asia and the Americas, leading to widespread outbreaks impacting millions of people. Despite more than 50 years of research into the pathogenesis of CHIKV, there is still no curative treatment available. Current management of CHIKV infections primarily involves providing supportive care to alleviate symptoms and improve the patient's quality of life. Given the ongoing threat of CHIKV, there is an urgent need to better understand its pathogenesis. This understanding is crucial for deciphering the mechanisms underlying the disease and for developing effective strategies for both prevention and management. This review aims to provide a comprehensive overview of CHIKV and its pathogenesis, shedding light on the complex interactions of viral genetics, host factors, immune responses, and vector-related factors. By exploring these intricate connections, the review seeks to contribute to the knowledge base surrounding CHIKV, offering insights that may ultimately lead to more effective prevention and management strategies for this re-emerging global health threat.
Collapse
Affiliation(s)
- Wesley Freppel
- Institute for Biomedicine and Glycomics, Gold Coast Campus, Griffith University, Southport, Australia
| | - Laurie A. Silva
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kenneth A. Stapleford
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Lara J. Herrero
- Institute for Biomedicine and Glycomics, Gold Coast Campus, Griffith University, Southport, Australia
| |
Collapse
|
3
|
Caillava AJ, Alfonso V, Tejerina Cibello M, Demaria MA, Coria LM, Cassataro J, Taboga OA, Alvarez DE. A vaccine candidate based on baculovirus displaying chikungunya virus E1-E2 envelope confers protection against challenge in mice. J Virol 2024; 98:e0101724. [PMID: 39440961 PMCID: PMC11575139 DOI: 10.1128/jvi.01017-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024] Open
Abstract
Chikungunya fever is a re-emerging mosquito-borne disease caused by the chikungunya virus (CHIKV) and produces acute arthritis that can progress to chronic disease with arthralgia. The first approved live-attenuated chikungunya vaccine has only recently become available for use in humans in the USA, but the access in endemic regions remains unmet. Here, we exploited the baculovirus display technology to develop a vectored vaccine candidate that exposes the CHIKV membrane proteins E1 and E2 on the baculovirus surface. Using recombinant baculovirus as vector vaccines has both productive and regulatory advantages: they are safe for handling and easy to produce in high titers and are non-pathogenic and non-replicative in mammals but have strong adjuvant properties by inducing humoral and cellular immune responses. CHIKV E1 and E2 envelope proteins with their own signal and transmembrane sequences were expressed on the surface of budded baculovirus virions. Immunization of C57BL/6 mice with non-adjuvanted recombinant baculovirus induced IgG antibodies against E2 with a predominant IgG2c subtype, neutralizing antibodies and a specific IFN-γ CD8+ T-cell response. Immunization with a second dose significantly boosted the antibody response, and mice immunized with two doses of the vaccine candidate were completely protected against challenge with CHIKV showing no detectable viremia or signs of disease. Altogether, baculovirus display of CHIKV envelope proteins served as an efficient vaccine platform against CHIKV.IMPORTANCEThe global spread of chikungunya virus (CHIKV) has disproportionately impacted the Americas that experienced a fourfold increase in 2023 in cases and deaths compared with the same period in 2022. The disease is characterized by acute fever and debilitating joint pain that can become chronic. Despite the socioeconomic burden related to the high morbidity rates of CHIKV infection, a vaccine for CHIKV is currently approved only in the USA. Vaccines are the most effective preventive measure against viral diseases, and advances in the development of different vaccine platforms such as nucleic acids and viral vectors have prompted the rapid deployment of vaccines to contain the COVID-19 pandemic. Here, we report the use of baculovirus display as a strategy for the design of a novel vaccine that provides sterilizing immunity in a mouse model of chikungunya disease. Our results encourage further research regarding the potential of baculovirus as platforms for human vaccine design.
Collapse
Affiliation(s)
- Ana J Caillava
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Victoria Alfonso
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), Instituto Nacional de Tecnología Agropecuaria (INTA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Malena Tejerina Cibello
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María Agostina Demaria
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Lorena M Coria
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín, San Martín, Argentina
| | - Juliana Cassataro
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín, San Martín, Argentina
| | - Oscar A Taboga
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), Instituto Nacional de Tecnología Agropecuaria (INTA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Diego E Alvarez
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín, San Martín, Argentina
| |
Collapse
|
4
|
Miao Q, Nguyen W, Zhu J, Liu G, van Oers MM, Tang B, Yan K, Larcher T, Suhrbier A, Pijlman GP. A getah virus-like-particle vaccine provides complete protection from viremia and arthritis in wild-type mice. Vaccine 2024; 42:126136. [PMID: 39004524 DOI: 10.1016/j.vaccine.2024.07.037] [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: 02/09/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
Getah virus (GETV) is an emerging mosquito-borne virus with economic impact on the livestock industry in East Asia. In this study, we successfully produced GETV virus-like particles (VLPs) in insect cells using the baculovirus expression vector system. We show that the GETV envelope glycoproteins were successfully expressed at the surface of the insect cell and were glycosylated. VLPs were isolated from the culture fluid as enveloped particles of 60-80 nm in diameter. Two 1 µg vaccinations with this GETV VLP vaccine, without adjuvant, generated neutralizing antibody responses and protected wild-type C57/BL6 mice against GETV viremia and arthritic disease. The GETV VLP vaccine may find application as a horse and/or pig vaccine in the future.
Collapse
Affiliation(s)
- Qiuhong Miao
- Laboratory of Virology, Wageningen University & Research, Wageningen, The Netherlands; Shanghai Veterinary Research Institute Chinese Academy of Agricultural Sciences, China.
| | - Wilson Nguyen
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia.
| | - Jie Zhu
- Shanghai Veterinary Research Institute Chinese Academy of Agricultural Sciences, China.
| | - Guangqing Liu
- Shanghai Veterinary Research Institute Chinese Academy of Agricultural Sciences, China.
| | - Monique M van Oers
- Laboratory of Virology, Wageningen University & Research, Wageningen, The Netherlands.
| | - Bing Tang
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia.
| | - Kexin Yan
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia.
| | | | - Andreas Suhrbier
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia; GVN Centre of Excellence, Australian Infectious Disease Research Centre, Brisbane, Queensland, Australia.
| | - Gorben P Pijlman
- Laboratory of Virology, Wageningen University & Research, Wageningen, The Netherlands.
| |
Collapse
|
5
|
Simo FBN, Burt FJ, Makoah NA. Chikungunya Virus Diagnosis: A Review of Current Antigen Detection Methods. Trop Med Infect Dis 2023; 8:365. [PMID: 37505661 PMCID: PMC10383795 DOI: 10.3390/tropicalmed8070365] [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: 05/15/2023] [Revised: 07/03/2023] [Accepted: 07/13/2023] [Indexed: 07/29/2023] Open
Abstract
Chikungunya is a mosquito-borne viral disease caused by the chikungunya virus (CHIKV). CHIKV is expanding at an alarming rate, potentially spreading and establishing endemicity in new areas where competent vectors are present. The dramatic spread of CHIKV in recent years highlights the urgent need to take precautionary measures and investigate options for control. It is crucial in developing nations where diagnostic tools are limited, and symptoms are similar to other prevalent diseases such as malaria and dengue. The most reliable method for diagnosing chikungunya virus is viral gene detection by RT-PCR. Alternative methods like detecting human antibody and viral antigen can also be used, especially in areas where resources are limited. In this review, we summarize the limited data on antigen detection immunoassays. We further explain the essential structural elements of the virus to help comprehend the scientific concepts underlying the testing methods, as well as future methods and diagnostic approaches under investigation.
Collapse
Affiliation(s)
- Fredy Brice Nemg Simo
- Division of Virology, Faculty of Health Sciences, University of The Free State, Bloemfontein 9301, Free State, South Africa
| | - Felicity Jane Burt
- Division of Virology, Faculty of Health Sciences, University of The Free State, Bloemfontein 9301, Free State, South Africa
- Division of Virology, National Health Laboratory Service, Bloemfontein 9301, Free State, South Africa
| | - Nigel Aminake Makoah
- Division of Virology, Faculty of Health Sciences, University of The Free State, Bloemfontein 9301, Free State, South Africa
| |
Collapse
|
6
|
Coirada FC, Fernandes ER, Mello LRD, Schuch V, Soares Campos G, Braconi CT, Boscardin SB, Santoro Rosa D. Heterologous DNA Prime- Subunit Protein Boost with Chikungunya Virus E2 Induces Neutralizing Antibodies and Cellular-Mediated Immunity. Int J Mol Sci 2023; 24:10517. [PMID: 37445695 DOI: 10.3390/ijms241310517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 07/15/2023] Open
Abstract
Chikungunya virus (CHIKV) has become a significant public health concern due to the increasing number of outbreaks worldwide and the associated comorbidities. Despite substantial efforts, there is no specific treatment or licensed vaccine against CHIKV to date. The E2 glycoprotein of CHIKV is a promising vaccine candidate as it is a major target of neutralizing antibodies during infection. In this study, we evaluated the immunogenicity of two DNA vaccines (a non-targeted and a dendritic cell-targeted vaccine) encoding a consensus sequence of E2CHIKV and a recombinant protein (E2*CHIKV). Mice were immunized with different homologous and heterologous DNAprime-E2* protein boost strategies, and the specific humoral and cellular immune responses were accessed. We found that mice immunized with heterologous non-targeted DNA prime- E2*CHIKV protein boost developed high levels of neutralizing antibodies, as well as specific IFN-γ producing cells and polyfunctional CD4+ and CD8+ T cells. We also identified 14 potential epitopes along the E2CHIKV protein. Furthermore, immunization with recombinant E2*CHIKV combined with the adjuvant AS03 presented the highest humoral response with neutralizing capacity. Finally, we show that the heterologous prime-boost strategy with the non-targeted pVAX-E2 DNA vaccine as the prime followed by E2* protein + AS03 boost is a promising combination to elicit a broad humoral and cellular immune response. Together, our data highlights the importance of E2CHIKV for the development of a CHIKV vaccine.
Collapse
Affiliation(s)
- Fernanda Caroline Coirada
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04023-062, Brazil
| | - Edgar Ruz Fernandes
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04023-062, Brazil
| | - Lucas Rodrigues de Mello
- Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04044-020, Brazil
| | - Viviane Schuch
- Departamento de Análises Clínicas e Toxicológicas, Universidade de São Paulo (USP), São Paulo 05508-000, Brazil
| | - Gúbio Soares Campos
- Laboratório de Virologia, Universidade Federal da Bahia (UFBA), Salvador 40110-909, Brazil
| | - Carla Torres Braconi
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04023-062, Brazil
| | - Silvia Beatriz Boscardin
- Departamento de Parasitologia, Universidade de São Paulo (USP), São Paulo 05508-000, Brazil
- Instituto Nacional de Ciência e Tecnologia de Investigação em Imunologia-INCT (III), São Paulo 05403-900, Brazil
| | - Daniela Santoro Rosa
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo 04023-062, Brazil
- Instituto Nacional de Ciência e Tecnologia de Investigação em Imunologia-INCT (III), São Paulo 05403-900, Brazil
| |
Collapse
|
7
|
Rao S, Abeyratne E, Freitas JR, Yang C, Tharmarajah K, Mostafavi H, Liu X, Zaman M, Mahalingam S, Zaid A, Taylor A. A booster regime of liposome-delivered live-attenuated CHIKV vaccine RNA genome protects against chikungunya virus disease in mice. Vaccine 2023; 41:3976-3988. [PMID: 37230889 DOI: 10.1016/j.vaccine.2023.05.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 05/27/2023]
Abstract
Mosquito-transmitted chikungunya virus (CHIKV) is the causal pathogen of CHIKV disease and is responsible for global epidemics of arthritic disease. CHIKV infection can lead to severe chronic and debilitating arthralgia, significantly impacting patient mobility and quality of life. Our previous studies have shown a live-attenuated CHIKV vaccine candidate, CHIKV-NoLS, to be effective in protecting against CHIKV disease in mice vaccinated with one dose. Further studies have demonstrated the value of a liposome RNA delivery system to deliver the RNA genome of CHIKV-NoLS directly in vivo, promoting de novo production of live-attenuated vaccine particles in vaccinated hosts. This system, designed to bypass live-attenuated vaccine production bottlenecks, uses CAF01 liposomes. However, one dose of CHIKV-NoLS CAF01 failed to provide systemic protection against CHIKV challenge in mice, with low levels of CHIKV-specific antibodies. Here we describe CHIKV-NoLS CAF01 booster vaccination regimes designed to increase vaccine efficacy. C57BL/6 mice were vaccinated with three doses of CHIKV-NoLS CAF01 either intramuscularly or subcutaneously. CHIKV-NoLS CAF01 vaccinated mice developed a systemic immune response against CHIKV that shared similarity to vaccination with CHIKV-NoLS, including high levels of CHIKV-specific neutralising antibodies in subcutaneously inoculated mice. CHIKV-NoLS CAF01 vaccinated mice were protected against disease signs and musculoskeletal inflammation when challenged with CHIKV. Mice given one dose of live-attenuated CHIKV-NoLS developed a long lasting protective immune response for up to 71 days. A clinically relevant CHIKV-NoLS CAF01 booster regime can overcome the challenges faced by our previous one dose strategy and provide systemic protection against CHIKV disease.
Collapse
Affiliation(s)
- Shambhavi Rao
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia
| | - Eranga Abeyratne
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia
| | - Joseph R Freitas
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia
| | - Chenying Yang
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia
| | - Kothila Tharmarajah
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia
| | - Helen Mostafavi
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia
| | - Xiang Liu
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia
| | - Mehfuz Zaman
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, 4222 Queensland, Australia
| | - Suresh Mahalingam
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia
| | - Ali Zaid
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia
| | - Adam Taylor
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia.
| |
Collapse
|
8
|
Varikkodan MM, Kunnathodi F, Azmi S, Wu TY. An Overview of Indian Biomedical Research on the Chikungunya Virus with Particular Reference to Its Vaccine, an Unmet Medical Need. Vaccines (Basel) 2023; 11:1102. [PMID: 37376491 DOI: 10.3390/vaccines11061102] [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: 04/30/2023] [Revised: 06/08/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Chikungunya virus (CHIKV) is an infectious agent spread by mosquitos, that has engendered endemic or epidemic outbreaks of Chikungunya fever (CHIKF) in Africa, South-East Asia, America, and a few European countries. Like most tropical infections, CHIKV is frequently misdiagnosed, underreported, and underestimated; it primarily affects areas with limited resources, like developing nations. Due to its high transmission rate and lack of a preventive vaccine or effective treatments, this virus poses a serious threat to humanity. After a 32-year hiatus, CHIKV reemerged as the most significant epidemic ever reported, in India in 2006. Since then, CHIKV-related research was begun in India, and up to now, more than 800 peer-reviewed research papers have been published by Indian researchers and medical practitioners. This review gives an overview of the outbreak history and CHIKV-related research in India, to favor novel high-quality research works intending to promote effective treatment and preventive strategies, including vaccine development, against CHIKV infection.
Collapse
Affiliation(s)
- Muhammed Muhsin Varikkodan
- Department of Bioscience Technology, College of Science, Chung Yuan Christian University, Chung-Li, Taoyuan City 320314, Taiwan
| | - Faisal Kunnathodi
- Scientific Research Center, Prince Sultan Military Medical City, Riyadh 11159, Saudi Arabia
| | - Sarfuddin Azmi
- Scientific Research Center, Prince Sultan Military Medical City, Riyadh 11159, Saudi Arabia
| | - Tzong-Yuan Wu
- Department of Bioscience Technology, College of Science, Chung Yuan Christian University, Chung-Li, Taoyuan City 320314, Taiwan
- R&D Center of Membrane Technology, Chung Yuan Christian University, Chung-Li, Taoyuan City 320314, Taiwan
| |
Collapse
|
9
|
Huang Z, Zhang Y, Li H, Zhu J, Song W, Chen K, Zhang Y, Lou Y. Vaccine development for mosquito-borne viral diseases. Front Immunol 2023; 14:1161149. [PMID: 37251387 PMCID: PMC10213220 DOI: 10.3389/fimmu.2023.1161149] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/13/2023] [Indexed: 05/31/2023] Open
Abstract
Mosquito-borne viral diseases are a group of viral illnesses that are predominantly transmitted by mosquitoes, including viruses from the Togaviridae and Flaviviridae families. In recent years, outbreaks caused by Dengue and Zika viruses from the Flaviviridae family, and Chikungunya virus from the Togaviridae family, have raised significant concerns for public health. However, there are currently no safe and effective vaccines available for these viruses, except for CYD-TDV, which has been licensed for Dengue virus. Efforts to control the transmission of COVID-19, such as home quarantine and travel restrictions, have somewhat limited the spread of mosquito-borne viral diseases. Several vaccine platforms, including inactivated vaccines, viral-vector vaccines, live attenuated vaccines, protein vaccines, and nucleic acid vaccines, are being developed to combat these viruses. This review analyzes the various vaccine platforms against Dengue, Zika, and Chikungunya viruses and provides valuable insights for responding to potential outbreaks.
Collapse
Affiliation(s)
- Zhiwei Huang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yuxuan Zhang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Hongyu Li
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jiajie Zhu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Wanchen Song
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Keda Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yanjun Zhang
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Yongliang Lou
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
10
|
Bhattacharjee S, Ghosh D, Saha R, Sarkar R, Kumar S, Khokhar M, Pandey RK. Mechanism of Immune Evasion in Mosquito-Borne Diseases. Pathogens 2023; 12:635. [PMID: 37242305 PMCID: PMC10222277 DOI: 10.3390/pathogens12050635] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
In recent decades, mosquito-borne illnesses have emerged as a major health burden in many tropical regions. These diseases, such as malaria, dengue fever, chikungunya, yellow fever, Zika virus infection, Rift Valley fever, Japanese encephalitis, and West Nile virus infection, are transmitted through the bite of infected mosquitoes. These pathogens have been shown to interfere with the host's immune system through adaptive and innate immune mechanisms, as well as the human circulatory system. Crucial immune checkpoints such as antigen presentation, T cell activation, differentiation, and proinflammatory response play a vital role in the host cell's response to pathogenic infection. Furthermore, these immune evasions have the potential to stimulate the human immune system, resulting in other associated non-communicable diseases. This review aims to advance our understanding of mosquito-borne diseases and the immune evasion mechanisms by associated pathogens. Moreover, it highlights the adverse outcomes of mosquito-borne disease.
Collapse
Affiliation(s)
| | - Debanjan Ghosh
- Department of Biotechnology, Pondicherry University, Puducherry 605014, India
| | - Rounak Saha
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry 605014, India
| | - Rima Sarkar
- DBT Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Saurav Kumar
- DBT Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Manoj Khokhar
- Department of Biochemistry, AIIMS, Jodhpur 342005, India
| | - Rajan Kumar Pandey
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Solna, Sweden
| |
Collapse
|
11
|
Novelo M, Dutra HLC, Metz HC, Jones MJ, Sigle LT, Frentiu FD, Allen SL, Chenoweth SF, McGraw EA. Dengue and chikungunya virus loads in the mosquito Aedes aegypti are determined by distinct genetic architectures. PLoS Pathog 2023; 19:e1011307. [PMID: 37043515 PMCID: PMC10124881 DOI: 10.1371/journal.ppat.1011307] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/24/2023] [Accepted: 03/19/2023] [Indexed: 04/13/2023] Open
Abstract
Aedes aegypti is the primary vector of the arboviruses dengue (DENV) and chikungunya (CHIKV). These viruses exhibit key differences in their vector interactions, the latter moving more quicky through the mosquito and triggering fewer standard antiviral pathways. As the global footprint of CHIKV continues to expand, we seek to better understand the mosquito's natural response to CHIKV-both to compare it to DENV:vector coevolutionary history and to identify potential targets in the mosquito for genetic modification. We used a modified full-sibling design to estimate the contribution of mosquito genetic variation to viral loads of both DENV and CHIKV. Heritabilities were significant, but higher for DENV (40%) than CHIKV (18%). Interestingly, there was no genetic correlation between DENV and CHIKV loads between siblings. These data suggest Ae. aegypti mosquitoes respond to the two viruses using distinct genetic mechanisms. We also examined genome-wide patterns of gene expression between High and Low CHIKV families representing the phenotypic extremes of viral load. Using RNAseq, we identified only two loci that consistently differentiated High and Low families: a long non-coding RNA that has been identified in mosquito screens post-infection and a distant member of a family of Salivary Gland Specific (SGS) genes. Interestingly, the latter gene is also associated with horizontal gene transfer between mosquitoes and the endosymbiotic bacterium Wolbachia. This work is the first to link the SGS gene to a mosquito phenotype. Understanding the molecular details of how this gene contributes to viral control in mosquitoes may, therefore, also shed light on its role in Wolbachia.
Collapse
Affiliation(s)
- Mario Novelo
- Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Center for Infectious Disease Dynamics, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Heverton LC Dutra
- Center for Infectious Disease Dynamics, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Hillery C. Metz
- Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Center for Infectious Disease Dynamics, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Matthew J. Jones
- Center for Infectious Disease Dynamics, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Leah T. Sigle
- Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Center for Infectious Disease Dynamics, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Francesca D. Frentiu
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Herston, Queensland, Australia
| | - Scott L. Allen
- School of Biological Sciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Stephen F. Chenoweth
- School of Biological Sciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Elizabeth A. McGraw
- Center for Infectious Disease Dynamics, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
12
|
Treffers EE, Tas A, Scholte FEM, de Ru AH, Snijder EJ, van Veelen PA, van Hemert MJ. The alphavirus nonstructural protein 2 NTPase induces a host translational shut-off through phosphorylation of eEF2 via cAMP-PKA-eEF2K signaling. PLoS Pathog 2023; 19:e1011179. [PMID: 36848386 PMCID: PMC9997916 DOI: 10.1371/journal.ppat.1011179] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 03/09/2023] [Accepted: 02/03/2023] [Indexed: 03/01/2023] Open
Abstract
Chikungunya virus (CHIKV) is a reemerging alphavirus. Since 2005, it has infected millions of people during outbreaks in Africa, Asia, and South/Central America. CHIKV replication depends on host cell factors at many levels and is expected to have a profound effect on cellular physiology. To obtain more insight into host responses to infection, stable isotope labeling with amino acids in cell culture and liquid chromatography-tandem mass spectrometry were used to assess temporal changes in the cellular phosphoproteome during CHIKV infection. Among the ~3,000 unique phosphorylation sites analyzed, the largest change in phosphorylation status was measured on residue T56 of eukaryotic elongation factor 2 (eEF2), which showed a >50-fold increase at 8 and 12 h p.i. Infection with other alphaviruses (Semliki Forest, Sindbis and Venezuelan equine encephalitis virus (VEEV)) triggered a similarly strong eEF2 phosphorylation. Expression of a truncated form of CHIKV or VEEV nsP2, containing only the N-terminal and NTPase/helicase domains (nsP2-NTD-Hel), sufficed to induce eEF2 phosphorylation, which could be prevented by mutating key residues in the Walker A and B motifs of the NTPase domain. Alphavirus infection or expression of nsP2-NTD-Hel resulted in decreased cellular ATP levels and increased cAMP levels. This did not occur when catalytically inactive NTPase mutants were expressed. The wild-type nsP2-NTD-Hel inhibited cellular translation independent of the C-terminal nsP2 domain, which was previously implicated in directing the virus-induced host shut-off for Old World alphaviruses. We hypothesize that the alphavirus NTPase activates a cellular adenylyl cyclase resulting in increased cAMP levels, thus activating PKA and subsequently eukaryotic elongation factor 2 kinase. This in turn triggers eEF2 phosphorylation and translational inhibition. We conclude that the nsP2-driven increase of cAMP levels contributes to the alphavirus-induced shut-off of cellular protein synthesis that is shared between Old and New World alphaviruses. MS Data are available via ProteomeXchange with identifier PXD009381.
Collapse
Affiliation(s)
- Emmely E. Treffers
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ali Tas
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Florine E. M. Scholte
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Arnoud H. de Ru
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Eric J. Snijder
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter A. van Veelen
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Martijn J. van Hemert
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
13
|
Shukla M, Chandley P, Tapryal S, Kumar N, Mukherjee SP, Rohatgi S. Expression, Purification, and Refolding of Chikungunya Virus Full-Length Envelope E2 Protein along with B-Cell and T-Cell Epitope Analyses Using Immuno-Informatics Approaches. ACS OMEGA 2022; 7:3491-3513. [PMID: 35128258 PMCID: PMC8811930 DOI: 10.1021/acsomega.1c05975] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/10/2021] [Indexed: 05/17/2023]
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus, which causes severe illness in humans and is responsible for epidemic outbreaks in Africa, Asia, North and South America, and Europe. Despite its increased global prevalence, no licensed vaccines are available to date for treating or preventing CHIKV infection. The envelope E2 protein is one of the promising subunit vaccine candidates against CHIKV. In this study, we describe successful cloning, expression, and purification of CHIKV E2 full-length (E2-FL) and truncated (E2-ΔC and E2-ΔNC) proteins in the Escherichia coli expression system. The recombinant E2 proteins were purified from inclusion bodies using Ni-NTA chromatography. Further, we describe a detailed refolding procedure for obtaining the CHIKV E2-FL protein in native conformation, which was confirmed using circular dichroism and Fourier transform infrared spectroscopy. BALB/c mice immunized with the three different E2 proteins exhibited increased E2-specific antibody titers compared to sham-immunized controls, suggesting induction of strong humoral immune response. On analyzing the E2-specific antibody response generated in immunized mice, the CHIKV E2-FL protein was observed to be the most immunogenic among the three different CHIKV E2 antigens used in the study. Our B-cell and T-cell epitope mapping results indicate that the presence of specific immunogenic peptides located in the N-terminal and C-terminal regions of the CHIKV E2-FL protein may contribute to its increased immunogenicity, compared to truncated CHIKV E2 proteins. In summary, our study provides a detailed protocol for expressing, purifying, and refolding of the CHIKV E2-FL protein and provides an understanding of its immunogenic epitopes, which can be exploited for the development of novel multiepitope-based anti-CHIKV vaccine strategies.
Collapse
Affiliation(s)
- Manisha Shukla
- Department
of Biosciences and Bioengineering, Indian
Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Pankaj Chandley
- Department
of Biosciences and Bioengineering, Indian
Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Suman Tapryal
- Department
of Biotechnology, Central University of
Rajasthan, Bandersindri,
Kishangarh, Ajmer 305817, Rajasthan, India
| | - Narendra Kumar
- Jaypee
University of Information Technology, Waknaghat, Solan 173234, India
| | - Sulakshana P. Mukherjee
- Department
of Biosciences and Bioengineering, Indian
Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Soma Rohatgi
- Department
of Biosciences and Bioengineering, Indian
Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| |
Collapse
|
14
|
Targovnik AM, Simonin JA, Mc Callum GJ, Smith I, Cuccovia Warlet FU, Nugnes MV, Miranda MV, Belaich MN. Solutions against emerging infectious and noninfectious human diseases through the application of baculovirus technologies. Appl Microbiol Biotechnol 2021; 105:8195-8226. [PMID: 34618205 PMCID: PMC8495437 DOI: 10.1007/s00253-021-11615-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/23/2022]
Abstract
Abstract
Baculoviruses are insect pathogens widely used as biotechnological tools in different fields of life sciences and technologies. The particular biology of these entities (biosafety viruses 1; large circular double-stranded DNA genomes, infective per se; generally of narrow host range on insect larvae; many of the latter being pests in agriculture) and the availability of molecular-biology procedures (e.g., genetic engineering to edit their genomes) and cellular resources (availability of cell lines that grow under in vitro culture conditions) have enabled the application of baculoviruses as active ingredients in pest control, as systems for the expression of recombinant proteins (Baculovirus Expression Vector Systems—BEVS) and as viral vectors for gene delivery in mammals or to display antigenic proteins (Baculoviruses applied on mammals—BacMam). Accordingly, BEVS and BacMam technologies have been introduced in academia because of their availability as commercial systems and ease of use and have also reached the human pharmaceutical industry, as incomparable tools in the development of biological products such as diagnostic kits, vaccines, protein therapies, and—though still in the conceptual stage involving animal models—gene therapies. Among all the baculovirus species, the Autographa californica multiple nucleopolyhedrovirus has been the most highly exploited in the above utilities for the human-biotechnology field. This review highlights the main achievements (in their different stages of development) of the use of BEVS and BacMam technologies for the generation of products for infectious and noninfectious human diseases. Key points • Baculoviruses can assist as biotechnological tools in human health problems. • Vaccines and diagnosis reagents produced in the baculovirus platform are described. • The use of recombinant baculovirus for gene therapy–based treatment is reviewed.
Collapse
Affiliation(s)
- Alexandra Marisa Targovnik
- Cátedra de Biotecnología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Buenos Aires, 1113, Argentina.
- Instituto de Nanobiotecnología (NANOBIOTEC), Facultad de Farmacia y Bioquímica, CONICET -Universidad de Buenos Aires, Junín 956, Sexto Piso, C1113AAD, 1113, Buenos Aires, Argentina.
| | - Jorge Alejandro Simonin
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular, Área Virosis de Insectos, Instituto de Microbiología Básica y Aplicada, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - Gregorio Juan Mc Callum
- Cátedra de Biotecnología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Buenos Aires, 1113, Argentina
- Instituto de Nanobiotecnología (NANOBIOTEC), Facultad de Farmacia y Bioquímica, CONICET -Universidad de Buenos Aires, Junín 956, Sexto Piso, C1113AAD, 1113, Buenos Aires, Argentina
| | - Ignacio Smith
- Cátedra de Biotecnología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Buenos Aires, 1113, Argentina
- Instituto de Nanobiotecnología (NANOBIOTEC), Facultad de Farmacia y Bioquímica, CONICET -Universidad de Buenos Aires, Junín 956, Sexto Piso, C1113AAD, 1113, Buenos Aires, Argentina
| | - Franco Uriel Cuccovia Warlet
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular, Área Virosis de Insectos, Instituto de Microbiología Básica y Aplicada, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - María Victoria Nugnes
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular, Área Virosis de Insectos, Instituto de Microbiología Básica y Aplicada, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - María Victoria Miranda
- Cátedra de Biotecnología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Buenos Aires, 1113, Argentina
- Instituto de Nanobiotecnología (NANOBIOTEC), Facultad de Farmacia y Bioquímica, CONICET -Universidad de Buenos Aires, Junín 956, Sexto Piso, C1113AAD, 1113, Buenos Aires, Argentina
| | - Mariano Nicolás Belaich
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular, Área Virosis de Insectos, Instituto de Microbiología Básica y Aplicada, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| |
Collapse
|
15
|
Constant LEC, Rajsfus BF, Carneiro PH, Sisnande T, Mohana-Borges R, Allonso D. Overview on Chikungunya Virus Infection: From Epidemiology to State-of-the-Art Experimental Models. Front Microbiol 2021; 12:744164. [PMID: 34675908 PMCID: PMC8524093 DOI: 10.3389/fmicb.2021.744164] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/07/2021] [Indexed: 12/27/2022] Open
Abstract
Chikungunya virus (CHIKV) is currently one of the most relevant arboviruses to public health. It is a member of the Togaviridae family and alphavirus genus and causes an arthritogenic disease known as chikungunya fever (CHIKF). It is characterized by a multifaceted disease, which is distinguished from other arbovirus infections by the intense and debilitating arthralgia that can last for months or years in some individuals. Despite the great social and economic burden caused by CHIKV infection, there is no vaccine or specific antiviral drugs currently available. Recent outbreaks have shown a change in the severity profile of the disease in which atypical and severe manifestation lead to hundreds of deaths, reinforcing the necessity to understand the replication and pathogenesis processes. CHIKF is a complex disease resultant from the infection of a plethora of cell types. Although there are several in vivo models for studying CHIKV infection, none of them reproduces integrally the disease signature observed in humans, which is a challenge for vaccine and drug development. Therefore, understanding the potentials and limitations of the state-of-the-art experimental models is imperative to advance in the field. In this context, the present review outlines the present knowledge on CHIKV epidemiology, replication, pathogenesis, and immunity and also brings a critical perspective on the current in vitro and in vivo state-of-the-art experimental models of CHIKF.
Collapse
Affiliation(s)
- Larissa E. C. Constant
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bia F. Rajsfus
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro H. Carneiro
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tháyna Sisnande
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ronaldo Mohana-Borges
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diego Allonso
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
16
|
Ma J, Wang H, Zheng X, Wu H, Yang S, Xia X. Western equine encephalitis virus virus-like particles from an insect cell-baculovirus system elicit the strong immune responses in mice. Biotechnol J 2021; 16:e2100008. [PMID: 34176228 DOI: 10.1002/biot.202100008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 11/07/2022]
Abstract
Western equine encephalitis virus (WEEV) causes lethal encephalitis in humans and equines, and it poses a serious public health threat in many countries. Therefore, the development of an efficient vaccine remains an important challenge for the prevention of WEEV infection. This study presents the first description of WEEV virus-like particles (VLPs) generated from insect cells using recombinant baculoviruses. WEEV VLPs with 206 adjuvant could trigger a strong cellular immune response; increase the levels of IL-2, IL-4 and IFN-γ; and induce a high level of neutralizing antibodies against WEEV in mice. These data showed that the insect cell-baculovirus system is suitable for the production of WEEV VLPs and that these VLPs could elicit the strong immunogenicity in mice. These results suggest a new, nonreplicating, and effective vaccine candidate against WEEV infection.
Collapse
Affiliation(s)
- JinZhu Ma
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China.,College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - HuaLei Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - XueXing Zheng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - HongXia Wu
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - SongTao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - XianZhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| |
Collapse
|
17
|
Torres-Ruesta A, Chee RSL, Ng LF. Insights into Antibody-Mediated Alphavirus Immunity and Vaccine Development Landscape. Microorganisms 2021; 9:microorganisms9050899. [PMID: 33922370 PMCID: PMC8145166 DOI: 10.3390/microorganisms9050899] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 12/11/2022] Open
Abstract
Alphaviruses are mosquito-borne pathogens distributed worldwide in tropical and temperate areas causing a wide range of symptoms ranging from inflammatory arthritis-like manifestations to the induction of encephalitis in humans. Historically, large outbreaks in susceptible populations have been recorded followed by the development of protective long-lasting antibody responses suggesting a potential advantageous role for a vaccine. Although the current understanding of alphavirus antibody-mediated immunity has been mainly gathered in natural and experimental settings of chikungunya virus (CHIKV) infection, little is known about the humoral responses triggered by other emerging alphaviruses. This knowledge is needed to improve serology-based diagnostic tests and the development of highly effective cross-protective vaccines. Here, we review the role of antibody-mediated immunity upon arthritogenic and neurotropic alphavirus infections, and the current research efforts for the development of vaccines as a tool to control future alphavirus outbreaks.
Collapse
Affiliation(s)
- Anthony Torres-Ruesta
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore; (A.T.-R.); (R.S.-L.C.)
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Rhonda Sin-Ling Chee
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore; (A.T.-R.); (R.S.-L.C.)
| | - Lisa F.P. Ng
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore; (A.T.-R.); (R.S.-L.C.)
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK
- Correspondence: ; Tel.: +65-6407-0028
| |
Collapse
|
18
|
Arthritogenic Alphavirus Capsid Protein. Life (Basel) 2021; 11:life11030230. [PMID: 33799673 PMCID: PMC7999773 DOI: 10.3390/life11030230] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 01/03/2023] Open
Abstract
In the past two decades Old World and arthritogenic alphavirus have been responsible for epidemics of polyarthritis, causing high morbidity and becoming a major public health concern. The multifunctional arthritogenic alphavirus capsid protein is crucial for viral infection. Capsid protein has roles in genome encapsulation, budding and virion assembly. Its role in multiple infection processes makes capsid protein an attractive target to exploit in combating alphaviral infection. In this review, we summarize the function of arthritogenic alphavirus capsid protein, and describe studies that have used capsid protein to develop novel arthritogenic alphavirus therapeutic and diagnostic strategies.
Collapse
|
19
|
Pijlman GP, Grose C, Hick TAH, Breukink HE, van den Braak R, Abbo SR, Geertsema C, van Oers MM, Martens DE, Esposito D. Relocation of the attTn7 Transgene Insertion Site in Bacmid DNA Enhances Baculovirus Genome Stability and Recombinant Protein Expression in Insect Cells. Viruses 2020; 12:v12121448. [PMID: 33339324 PMCID: PMC7765880 DOI: 10.3390/v12121448] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/09/2020] [Accepted: 12/14/2020] [Indexed: 01/22/2023] Open
Abstract
Baculovirus expression vectors are successfully used for the commercial production of complex (glyco)proteins in eukaryotic cells. The genome engineering of single-copy baculovirus infectious clones (bacmids) in E. coli has been valuable in the study of baculovirus biology, but bacmids are not yet widely applied as expression vectors. An important limitation of first-generation bacmids for large-scale protein production is the rapid loss of gene of interest (GOI) expression. The instability is caused by the mini-F replicon in the bacmid backbone, which is non-essential for baculovirus replication in insect cells, and carries the adjacent GOI in between attTn7 transposition sites. In this paper, we test the hypothesis that relocation of the attTn7 transgene insertion site away from the mini-F replicon prevents deletion of the GOI, thereby resulting in higher and prolonged recombinant protein expression levels. We applied lambda red genome engineering combined with SacB counterselection to generate a series of bacmids with relocated attTn7 sites and tested their performance by comparing the relative expression levels of different GOIs. We conclude that GOI expression from the odv-e56 (pif-5) locus results in higher overall expression levels and is more stable over serial passages compared to the original bacmid. Finally, we evaluated this improved next-generation bacmid during a bioreactor scale-up of Sf9 insect cells in suspension to produce enveloped chikungunya virus-like particles as a model vaccine.
Collapse
Affiliation(s)
- Gorben P. Pijlman
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708PB Wageningen, The Netherlands; (T.A.H.H.); (H.E.B.); (R.v.d.B.); (S.R.A.); (C.G.); (M.M.v.O.)
- Correspondence: ; Tel.: +31-317-484498
| | - Carissa Grose
- Protein Expression Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. PO Box B, Frederick, MD 21702, USA; (C.G.); (D.E.)
| | - Tessy A. H. Hick
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708PB Wageningen, The Netherlands; (T.A.H.H.); (H.E.B.); (R.v.d.B.); (S.R.A.); (C.G.); (M.M.v.O.)
| | - Herman E. Breukink
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708PB Wageningen, The Netherlands; (T.A.H.H.); (H.E.B.); (R.v.d.B.); (S.R.A.); (C.G.); (M.M.v.O.)
| | - Robin van den Braak
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708PB Wageningen, The Netherlands; (T.A.H.H.); (H.E.B.); (R.v.d.B.); (S.R.A.); (C.G.); (M.M.v.O.)
| | - Sandra R. Abbo
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708PB Wageningen, The Netherlands; (T.A.H.H.); (H.E.B.); (R.v.d.B.); (S.R.A.); (C.G.); (M.M.v.O.)
| | - Corinne Geertsema
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708PB Wageningen, The Netherlands; (T.A.H.H.); (H.E.B.); (R.v.d.B.); (S.R.A.); (C.G.); (M.M.v.O.)
| | - Monique M. van Oers
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708PB Wageningen, The Netherlands; (T.A.H.H.); (H.E.B.); (R.v.d.B.); (S.R.A.); (C.G.); (M.M.v.O.)
| | - Dirk E. Martens
- Bioprocess Engineering, Wageningen University, Droevendaalsesteeg 1, 6708PB Wageningen, The Netherlands;
| | - Dominic Esposito
- Protein Expression Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc. PO Box B, Frederick, MD 21702, USA; (C.G.); (D.E.)
| |
Collapse
|
20
|
Laureti M, Paradkar PN, Fazakerley JK, Rodriguez-Andres J. Superinfection Exclusion in Mosquitoes and Its Potential as an Arbovirus Control Strategy. Viruses 2020; 12:v12111259. [PMID: 33167513 PMCID: PMC7694488 DOI: 10.3390/v12111259] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 12/19/2022] Open
Abstract
The continuing emergence of arbovirus disease outbreaks around the world, despite the use of vector control strategies, warrants the development of new strategies to reduce arbovirus transmission. Superinfection exclusion, a phenomenon whereby a primary virus infection prevents the replication of a second closely related virus, has potential to control arbovirus disease emergence and outbreaks. This phenomenon has been observed for many years in plants, insects and mammalian cells. In this review, we discuss the significance of identifying novel vector control strategies, summarize studies exploring arbovirus superinfection exclusion and consider the potential for this phenomenon to be the basis for novel arbovirus control strategies.
Collapse
Affiliation(s)
- Mathilde Laureti
- Peter Doherty Institute for Infection and Immunity and Faculty of Veterinary and Agricultural Sciences, University of Melbourne, VIC 3000 Melbourne, Australia;
- CSIRO Health & Biosecurity, Australian Centre for Diseases Preparedness, VIC 3220 Geelong, Australia;
- Correspondence: (M.L.); (J.R.-A.)
| | - Prasad N. Paradkar
- CSIRO Health & Biosecurity, Australian Centre for Diseases Preparedness, VIC 3220 Geelong, Australia;
| | - John K. Fazakerley
- Peter Doherty Institute for Infection and Immunity and Faculty of Veterinary and Agricultural Sciences, University of Melbourne, VIC 3000 Melbourne, Australia;
| | - Julio Rodriguez-Andres
- Peter Doherty Institute for Infection and Immunity and Faculty of Veterinary and Agricultural Sciences, University of Melbourne, VIC 3000 Melbourne, Australia;
- Correspondence: (M.L.); (J.R.-A.)
| |
Collapse
|
21
|
Kumar S, Singh VK, Vasam M, Patil PS, Dhaked RK, Ansari AS, Lohiya NK, Parashar D, Tapryal S. An in vitro refolding method to produce oligomers of anti-CHIKV, E2-IgM Fc fusion subunit vaccine candidates expressed in E. coli. J Immunol Methods 2020; 487:112869. [PMID: 32971119 DOI: 10.1016/j.jim.2020.112869] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 09/12/2020] [Accepted: 09/16/2020] [Indexed: 12/25/2022]
Abstract
Recombinant envelope protein-1 (E1) and E2 of Chikungunya virus (CHIKV) has been shown to elicit neutralizing antibodies and a balanced Th1/Th2 response in mice however with limited protection. Recently reported CHIK virus-like particles showed augmented immunity and protection in adult mice in comparison to E1 and E2, however exacerbated the disease in aged subjects. In order to improve the overall efficacy of protein based vaccines, novel strategies need to be adopted. The discovery of IgM Fc receptor (FcμR) and its role in humoral immune response led us to hypothesise that fusion of an antigen with Fc of IgM may enhance its immunogenicity by polymerizing it and FcμR mediated activation of B and other immune cells. We report in the current study, expression of E2 subunit of CHIKV in fusion with various IgM Fc domains/peptides in E. coli, their in-vitro refolding, characterization and immune response in C57BL/6 mice. Candidates fused with CH3-CH4 Fc fragment produced stable oligomers, whereas the one fused with peptides remained monomeric. The latter elicited a strong humoral and a balanced Th1/Th2 response in mice, whereas the polymeric candidate despite eliciting a strong humoral response, stimulated a biased Th1 response and exhibited higher virus neutralization in Vero cells.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Vikas Kumar Singh
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Manohar Vasam
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Poonam Shewale Patil
- Dengue & Chikungunya Group, ICMR-National Institute of Virology, 20-A Dr. Ambedkar Road, Pune 411001, India
| | - Rajeev K Dhaked
- Department of Zoology, Centre for Advanced Studies, University of Rajasthan, JLN Marg, Jaipur, Rajasthan 302004, India
| | - Abdul S Ansari
- Department of Zoology, Centre for Advanced Studies, University of Rajasthan, JLN Marg, Jaipur, Rajasthan 302004, India
| | - Nirmal K Lohiya
- Department of Zoology, Centre for Advanced Studies, Indian Society for the Study of Reproduction & Fertility, University of Rajasthan, Jaipur, Rajasthan 302004, India
| | - Deepti Parashar
- Dengue & Chikungunya Group, ICMR-National Institute of Virology, 20-A Dr. Ambedkar Road, Pune 411001, India
| | - Suman Tapryal
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Ajmer, Rajasthan 305817, India.
| |
Collapse
|
22
|
Bagno FF, Godói LC, Figueiredo MM, Sérgio SAR, Moraes TDFS, Salazar NDC, Kim YC, Reyes-Sandoval A, da Fonseca FG. Chikungunya E2 Protein Produced in E. coli and HEK293-T Cells-Comparison of Their Performances in ELISA. Viruses 2020; 12:E939. [PMID: 32858804 PMCID: PMC7552038 DOI: 10.3390/v12090939] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/29/2022] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne pathogen that causes a disease characterized by the acute onset of fever accompanied by arthralgia and intense joint pain. Clinical similarities and cocirculation of this and other arboviruses in many tropical countries highlight the necessity for efficient and accessible diagnostic tools. CHIKV envelope proteins are highly conserved among alphaviruses and, particularly, the envelope 2 glycoprotein (CHIKV-E2) appears to be immunodominant and has a considerable serodiagnosis potential. Here, we investigate how glycosylation of CHIKV-E2 affects antigen/antibody interaction and how this affects the performance of CHIKV-E2-based Indirect ELISA tests. We compare two CHIKV-E2 recombinant antigens produced in different expression systems: prokaryotic-versus eukaryotic-made recombinant proteins. CHIKV-E2 antigens are expressed either in E. coli BL21(DE3)-a prokaryotic system unable to produce post-translational modifications-or in HEK-293T mammalian cells-a eukaryotic system able to add post-translational modifications, including glycosylation sites. Both prokaryotic and eukaryotic recombinant CHIKV-E2 react strongly to anti-CHIKV IgG antibodies, showing accuracy levels that are higher than 90%. However, the glycan-added viral antigen presents better sensitivity and specificity (85 and 98%) than the non-glycosylated antigen (81 and 71%, respectively) in anti-CHIKV IgM ELISA assays.
Collapse
Affiliation(s)
- Flávia Fonseca Bagno
- Centro de Tecnologia em Vacinas (CT-Vacinas), Parque Tecnológico da UFMG (BH-Tec), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG 31320-000, Brazil; (F.F.B.); (L.C.G.); (M.M.F.); (S.A.R.S.); (T.d.F.S.M.); (N.d.C.S.)
- Laboratório de Virologia Molecular e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas (ICB/UFMG), Belo Horizonte-MG 31270-901, Brazil
| | - Lara Carvalho Godói
- Centro de Tecnologia em Vacinas (CT-Vacinas), Parque Tecnológico da UFMG (BH-Tec), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG 31320-000, Brazil; (F.F.B.); (L.C.G.); (M.M.F.); (S.A.R.S.); (T.d.F.S.M.); (N.d.C.S.)
- Colégio Técnico da Universidade Federal de Minas Gerais (COLTEC), Belo Horizonte-MG 31270-901, Brazil
| | - Maria Marta Figueiredo
- Centro de Tecnologia em Vacinas (CT-Vacinas), Parque Tecnológico da UFMG (BH-Tec), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG 31320-000, Brazil; (F.F.B.); (L.C.G.); (M.M.F.); (S.A.R.S.); (T.d.F.S.M.); (N.d.C.S.)
| | - Sarah Aparecida Rodrigues Sérgio
- Centro de Tecnologia em Vacinas (CT-Vacinas), Parque Tecnológico da UFMG (BH-Tec), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG 31320-000, Brazil; (F.F.B.); (L.C.G.); (M.M.F.); (S.A.R.S.); (T.d.F.S.M.); (N.d.C.S.)
| | - Thaís de Fátima Silva Moraes
- Centro de Tecnologia em Vacinas (CT-Vacinas), Parque Tecnológico da UFMG (BH-Tec), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG 31320-000, Brazil; (F.F.B.); (L.C.G.); (M.M.F.); (S.A.R.S.); (T.d.F.S.M.); (N.d.C.S.)
- Laboratório de Virologia Molecular e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas (ICB/UFMG), Belo Horizonte-MG 31270-901, Brazil
| | - Natália de Castro Salazar
- Centro de Tecnologia em Vacinas (CT-Vacinas), Parque Tecnológico da UFMG (BH-Tec), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG 31320-000, Brazil; (F.F.B.); (L.C.G.); (M.M.F.); (S.A.R.S.); (T.d.F.S.M.); (N.d.C.S.)
| | - Young Chan Kim
- The Jenner Institute, Nuffield Department of Medicine, The Henry Wellcome Building for Molecular Physiology, Roosevelt Drive, University of Oxford, Oxford OX3 7DQ, UK; (Y.C.K.); (A.R.-S.)
| | - Arturo Reyes-Sandoval
- The Jenner Institute, Nuffield Department of Medicine, The Henry Wellcome Building for Molecular Physiology, Roosevelt Drive, University of Oxford, Oxford OX3 7DQ, UK; (Y.C.K.); (A.R.-S.)
| | - Flávio Guimarães da Fonseca
- Centro de Tecnologia em Vacinas (CT-Vacinas), Parque Tecnológico da UFMG (BH-Tec), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG 31320-000, Brazil; (F.F.B.); (L.C.G.); (M.M.F.); (S.A.R.S.); (T.d.F.S.M.); (N.d.C.S.)
- Laboratório de Virologia Molecular e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas (ICB/UFMG), Belo Horizonte-MG 31270-901, Brazil
| |
Collapse
|
23
|
Schrauf S, Tschismarov R, Tauber E, Ramsauer K. Current Efforts in the Development of Vaccines for the Prevention of Zika and Chikungunya Virus Infections. Front Immunol 2020; 11:592. [PMID: 32373111 PMCID: PMC7179680 DOI: 10.3389/fimmu.2020.00592] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 03/13/2020] [Indexed: 01/07/2023] Open
Abstract
Arboviruses represent major challenges to public health, particularly in tropical, and subtropical regions, and a substantial risk to other parts of the world as respective vectors extend their habitats. In recent years, two viruses transmitted by Aedes mosquitoes, Chikungunya and Zika virus, have gathered increased interest. After decades of regionally constrained outbreaks, both viruses have recently caused explosive outbreaks on an unprecedented scale, causing immense suffering and massive economic burdens in affected regions. Chikungunya virus causes an acute febrile illness that often transitions into a chronic manifestation characterized by debilitating arthralgia and/or arthritis in a substantial subset of infected individuals. Zika infection frequently presents as a mild influenza-like illness, often subclinical, but can cause severe complications such as congenital malformations in pregnancy and neurological disorders, including Guillain-Barré syndrome. With no specific treatments or vaccines available, vector control remains the most effective measure to manage spread of these diseases. Given that both viruses cause antibody responses that confer long-term, possibly lifelong protection and that such responses are cross-protective against the various circulating genetic lineages, the development of Zika and Chikungunya vaccines represents a promising route for disease control. In this review we provide a brief overview on Zika and Chikungunya viruses, the etiology and epidemiology of the illnesses they cause and the host immune response against them, before summarizing past and current efforts to develop vaccines to alleviate the burden caused by these emerging diseases. The development of the urgently needed vaccines is hampered by several factors including the unpredictable epidemiology, feasibility of rapid clinical trial implementation during outbreaks and regulatory pathways. We will give an overview of the current developments.
Collapse
|
24
|
Gao S, Song S, Zhang L. Recent Progress in Vaccine Development Against Chikungunya Virus. Front Microbiol 2019; 10:2881. [PMID: 31921059 PMCID: PMC6930866 DOI: 10.3389/fmicb.2019.02881] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 11/29/2019] [Indexed: 01/25/2023] Open
Abstract
Chikungunya fever (CHIKF) is an acute infectious disease that is mediated by the mosquito-transmitted chikungunya virus (CHIKV). People infected with CHIKV may experience high fever, severe joint pain, skin rash, and headache. In recent years, this disease has become a global public health problem. However, there is no licensed vaccine available for CHIKV. Accumulating research data have provided novel approaches and new directions for the development of CHIKV vaccines. Our review focuses on recent progress in CHIKV vaccine studies. The potential vaccine candidates are classified into seven types: inactivated vaccine, subunit vaccine, live-attenuated vaccine, recombinant virus-vectored vaccine, virus-like particle vaccine, chimeric vaccine, and nucleic acid vaccine. These studies will provide important insights into the future development of CHIKV vaccines.
Collapse
Affiliation(s)
- Shan Gao
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Siqi Song
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.,School of Basic Medicine, Qingdao University, Qingdao, China
| | - Leiliang Zhang
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
25
|
Highly efficient production of rabies virus glycoprotein G ectodomain in Sf9 insect cells. 3 Biotech 2019; 9:385. [PMID: 31656723 DOI: 10.1007/s13205-019-1920-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 09/24/2019] [Indexed: 10/25/2022] Open
Abstract
In the present study, we developed a complete process to produce in insect cells a high amount of the ectodomain of rabies virus glycoprotein G (GE) as suitable antigen for detecting anti-rabies antibodies. Using the baculovirus expression vector system in Sf9 insect cells combined with a novel chimeric promoter (polh-pSeL), the expression level reached a yield of 4.1 ± 0.3 mg/L culture, which was significantly higher than that achieved with the standard polh promoter alone. The protein was recovered from the cell lysates and easily purified in only one step by metal ion affinity chromatography, with a yield of 95% and a purity of 87%. Finally, GE was successfully used in an assay to detect specific antibodies in serum samples derived from rabies-vaccinated animals. The efficient strategy developed in this work is an interesting method to produce high amounts of this glycoprotein.
Collapse
|
26
|
Carpentier KS, Davenport BJ, Haist KC, McCarthy MK, May NA, Robison A, Ruckert C, Ebel GD, Morrison TE. Discrete viral E2 lysine residues and scavenger receptor MARCO are required for clearance of circulating alphaviruses. eLife 2019; 8:e49163. [PMID: 31596239 PMCID: PMC6839921 DOI: 10.7554/elife.49163] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 10/04/2019] [Indexed: 12/12/2022] Open
Abstract
The magnitude and duration of vertebrate viremia is a critical determinant of arbovirus transmission, geographic spread, and disease severity. We find that multiple alphaviruses, including chikungunya (CHIKV), Ross River (RRV), and o'nyong 'nyong (ONNV) viruses, are cleared from the circulation of mice by liver Kupffer cells, impeding viral dissemination. Clearance from the circulation was independent of natural antibodies or complement factor C3, and instead relied on scavenger receptor SR-A6 (MARCO). Remarkably, lysine to arginine substitutions at distinct residues within the E2 glycoproteins of CHIKV and ONNV (E2 K200R) as well as RRV (E2 K251R) allowed for escape from clearance and enhanced viremia and dissemination. Mutational analysis revealed that viral clearance from the circulation is strictly dependent on the presence of lysine at these positions. These findings reveal a previously unrecognized innate immune pathway that controls alphavirus viremia and dissemination in vertebrate hosts, ultimately influencing disease severity and likely transmission efficiency.
Collapse
Affiliation(s)
- Kathryn S Carpentier
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Bennett J Davenport
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Kelsey C Haist
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Mary K McCarthy
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Nicholas A May
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| | - Alexis Robison
- Department of Microbiology, Immunology, and PathologyColorado State UniversityFort CollinsUnited States
| | - Claudia Ruckert
- Department of Microbiology, Immunology, and PathologyColorado State UniversityFort CollinsUnited States
| | - Gregory D Ebel
- Department of Microbiology, Immunology, and PathologyColorado State UniversityFort CollinsUnited States
| | - Thomas E Morrison
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraUnited States
| |
Collapse
|
27
|
Expression, purification and functional characterization of recombinant hypervariable region (HVR) of Chikungunya virus nsP3 protein. 3 Biotech 2019; 9:235. [PMID: 31139550 DOI: 10.1007/s13205-019-1759-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 05/13/2019] [Indexed: 12/23/2022] Open
Abstract
One of the most important rapidly emerging mosquito-borne alphavirus is Chikungunya virus (CHIKV). There is a necessity to develop anti-CHIKV therapeutics, as neither antiviral drug nor vaccines have been licensed yet. Several CHIKV proteins are being studied worldwide, but non-structural protein 3 (nsP3) has been less explored. This protein consists of three domains: macrodomain, alphavirus unique domain (AUD) and hypervariable region (HVR). The proline-rich regions of HVR contain SRC homology 3 (SH3)-binding domain which is essential for its functionality. Interaction of these motifs with host amphiphysin protein is crucial for viral RNA replication. Restricting the interactions of HVR could lead to inhibition of viral life cycle. Therefore, the present study focuses on purification of HVR protein and its structural and functional assay for therapeutic intervention in future use. In order to obtain purified protein, HVR region was amplified from TOPO clones of nsP3 of IND-06-Guj strain and cloned into expression vector. Expression and solubilization of the protein were optimized at various conditions of salt, detergent and imidazole before purification. The soluble recombinant HVR (His-HVR) protein was purified using affinity chromatography. Purified protein was analyzed for structural studies and functional assays. Circular dichroism of His-HVR protein was performed for structural study, and it was observed that it consists of mostly random coils. For functional assay, co-pull down of His-HVR protein was performed with endogenous amphiphysin-I protein of N2a cells and was analyzed using Western blotting. This purified protein obtained could be used as a potential target reagent for novel therapeutic interventions in the future.
Collapse
|
28
|
Göertz GP, Lingemann M, Geertsema C, Abma-Henkens MHC, Vogels CBF, Koenraadt CJM, van Oers MM, Pijlman GP. Conserved motifs in the hypervariable domain of chikungunya virus nsP3 required for transmission by Aedes aegypti mosquitoes. PLoS Negl Trop Dis 2018; 12:e0006958. [PMID: 30412583 PMCID: PMC6249005 DOI: 10.1371/journal.pntd.0006958] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/21/2018] [Accepted: 10/29/2018] [Indexed: 01/09/2023] Open
Abstract
Background Chikungunya virus (CHIKV) is a re-emerging arthropod-borne (arbo)virus that causes chikungunya fever in humans and is predominantly transmitted by Aedes aegypti mosquitoes. The CHIKV replication machinery consists of four non-structural proteins (nsP1-4) that additionally require the presence of a number of host proteins for replication of the viral RNA. NsP3 is essential for CHIKV replication and has a conserved macro, central and C-terminal hypervariable domain (HVD). The HVD is intrinsically disordered and interacts with various host proteins via conserved short peptide motifs: A proline-rich (P-rich) motif that has affinity for SH3-domain containing proteins and duplicate FGDF motifs with affinity for G3BP and its mosquito homologue Rasputin. The importance of these motifs for infection of mammalian cells has previously been implicated. However, their role during CHIKV infection of mosquito cells and transmission by mosquitoes remains unclear. Methodology / Principal findings Here, we show that in-frame deletion of the P-rich motif is lethal for CHIKV replication in both mosquito and mammalian cells. However, while mutagenesis of the P-rich motif negatively affects replication both in mammalian and mosquito cells, it did not compromise the infection and transmission of CHIKV by Ae. aegypti mosquitoes. Mutagenesis of both FGDF motifs together completely inactivated CHIKV replication in both mammalian and mosquito cells. Importantly, mutation of a single FGDF motif attenuated CHIKV replication in mammalian cells, while replication in mosquito cells was similar to wild type. Surprisingly, CHIKV mutants containing only a single FGDF motif were efficiently transmitted by Ae. aegypti. Conclusions / Significance The P-rich motif in CHIKV nsP3 is dispensable for transmission by mosquitoes. A single FGDF motif is sufficient for infection and dissemination in mosquitoes, but duplicate FGDF motifs are required for the efficient infection from the mosquito saliva to a vertebrate host. These results contribute to understanding the dynamics of the alphavirus transmission cycle and may help the development of arboviral intervention strategies. Chikungunya virus (CHIKV) is a re-emerging arthropod-borne virus that is transmitted predominantly by Aedes aegypti mosquitoes. In 2016 alone CHIKV caused over 100.000 infections in South-America, exemplifying the impact of CHIKV disease. Previous research has suggested that the CHIKV non-structural protein 3 (nsP3) may determine the infection of mosquitoes. NsP3 is known to interact with several host proteins through a conserved proline (P)-rich and duplicate FGDF motifs that are present in its C-terminal domain. Here we investigated the importance of these conserved motifs for the infection and replication of CHIKV in both Aedes mosquito cells and mammalian cells. Furthermore, we assessed the role of these motifs for the transmission by Ae. aegypti mosquitoes via infectious bloodmeal experiments. We show that mutation of the P-rich motif negatively affects the replication of CHIKV in both mammalian and mosquito cells. In contrast, mutating the P-rich motif did not affect the transmission by Ae. aegypti. Mutation of both FGDF motifs together completely inactivated CHIKV in mammalian and mosquito cells, while mutation of a single FGDF motif negatively affected replication only in mammalian cells. Importantly, CHIKV containing only a single FGDF motif was still efficiently transmitted by Ae. aegypti mosquitoes. These results contribute to understanding the key interactions between alphaviruses and their mosquito vector.
Collapse
Affiliation(s)
- Giel P. Göertz
- Laboratory of Virology, Wageningen University & Research, PB, Wageningen, The Netherlands
| | - Marit Lingemann
- Laboratory of Virology, Wageningen University & Research, PB, Wageningen, The Netherlands
| | - Corinne Geertsema
- Laboratory of Virology, Wageningen University & Research, PB, Wageningen, The Netherlands
| | | | - Chantal B. F. Vogels
- Laboratory of Entomology, Wageningen University & Research, PB, Wageningen, The Netherlands
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, United States of America
| | | | - Monique M. van Oers
- Laboratory of Virology, Wageningen University & Research, PB, Wageningen, The Netherlands
| | - Gorben P. Pijlman
- Laboratory of Virology, Wageningen University & Research, PB, Wageningen, The Netherlands
- * E-mail:
| |
Collapse
|
29
|
Tanabe ISB, Tanabe ELL, Santos EC, Martins WV, Araújo IMTC, Cavalcante MCA, Lima ARV, Câmara NOS, Anderson L, Yunusov D, Bassi ÊJ. Cellular and Molecular Immune Response to Chikungunya Virus Infection. Front Cell Infect Microbiol 2018; 8:345. [PMID: 30364124 PMCID: PMC6191487 DOI: 10.3389/fcimb.2018.00345] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 09/11/2018] [Indexed: 11/13/2022] Open
Abstract
Chikungunya virus (CHIKV) is a re-emergent arthropod-borne virus (arbovirus) that causes a disease characterized primarily by fever, rash and severe persistent polyarthralgia. In the last decade, CHIKV has become a serious public health problem causing several outbreaks around the world. Despite the fact that CHIKV has been around since 1952, our knowledge about immunopathology, innate and adaptive immune response involved in this infectious disease is incomplete. In this review, we provide an updated summary of the current knowledge about immune response to CHIKV and about soluble immunological markers associated with the morbidity, prognosis and chronicity of this arbovirus disease. In addition, we discuss the progress in the research of new vaccines for preventing CHIKV infection and the use of monoclonal antibodies as a promising therapeutic strategy.
Collapse
Affiliation(s)
- Ithallo S B Tanabe
- IMUNOREG-Grupo de Pesquisa em Regulação da Resposta Imune, Laboratório de Pesquisas em Virologia e Imunologia, Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Maceió, Brazil
| | - Eloiza L L Tanabe
- IMUNOREG-Grupo de Pesquisa em Regulação da Resposta Imune, Laboratório de Pesquisas em Virologia e Imunologia, Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Maceió, Brazil
| | - Elane C Santos
- IMUNOREG-Grupo de Pesquisa em Regulação da Resposta Imune, Laboratório de Pesquisas em Virologia e Imunologia, Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Maceió, Brazil
| | - Wanessa V Martins
- IMUNOREG-Grupo de Pesquisa em Regulação da Resposta Imune, Laboratório de Pesquisas em Virologia e Imunologia, Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Maceió, Brazil
| | - Isadora M T C Araújo
- IMUNOREG-Grupo de Pesquisa em Regulação da Resposta Imune, Laboratório de Pesquisas em Virologia e Imunologia, Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Maceió, Brazil
| | - Maria C A Cavalcante
- IMUNOREG-Grupo de Pesquisa em Regulação da Resposta Imune, Laboratório de Pesquisas em Virologia e Imunologia, Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Maceió, Brazil
| | - Ana R V Lima
- IMUNOREG-Grupo de Pesquisa em Regulação da Resposta Imune, Laboratório de Pesquisas em Virologia e Imunologia, Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Maceió, Brazil
| | - Niels O S Câmara
- Laboratório de Imunobiologia dos Transplantes, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Leticia Anderson
- IMUNOREG-Grupo de Pesquisa em Regulação da Resposta Imune, Laboratório de Pesquisas em Virologia e Imunologia, Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Maceió, Brazil.,Centro Universitário CESMAC, Maceió, Brazil
| | - Dinar Yunusov
- Cold Spring Harbor Laboratory, Genome Research Center, Woodbury, NY, United States
| | - Ênio J Bassi
- IMUNOREG-Grupo de Pesquisa em Regulação da Resposta Imune, Laboratório de Pesquisas em Virologia e Imunologia, Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Alagoas, Maceió, Brazil
| |
Collapse
|
30
|
Wong KZ, Chu JJH. The Interplay of Viral and Host Factors in Chikungunya Virus Infection: Targets for Antiviral Strategies. Viruses 2018; 10:E294. [PMID: 29849008 PMCID: PMC6024654 DOI: 10.3390/v10060294] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/13/2018] [Accepted: 05/28/2018] [Indexed: 12/14/2022] Open
Abstract
Chikungunya virus (CHIKV) has re-emerged as one of the many medically important arboviruses that have spread rampantly across the world in the past decade. Infected patients come down with acute fever and rashes, and a portion of them suffer from both acute and chronic arthralgia. Currently, there are no targeted therapeutics against this debilitating virus. One approach to develop potential therapeutics is by understanding the viral-host interactions. However, to date, there has been limited research undertaken in this area. In this review, we attempt to briefly describe and update the functions of the different CHIKV proteins and their respective interacting host partners. In addition, we also survey the literature for other reported host factors and pathways involved during CHIKV infection. There is a pressing need for an in-depth understanding of the interaction between the host environment and CHIKV in order to generate potential therapeutics.
Collapse
Affiliation(s)
- Kai Zhi Wong
- Laboratory of Molecular RNA Virology & Antiviral Strategies, Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University Health System, 5 Science Drive 2, National University of Singapore, Singapore 117597, Singapore.
| | - Justin Jang Hann Chu
- Laboratory of Molecular RNA Virology & Antiviral Strategies, Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University Health System, 5 Science Drive 2, National University of Singapore, Singapore 117597, Singapore.
- Institute of Molecular & Cell Biology, Agency for Science, Technology & Research (A*STAR), 61 Biopolis Drive, Proteos #06-05, Singapore 138673, Singapore.
| |
Collapse
|
31
|
Singh A, Kumar A, Yadav R, Uversky VN, Giri R. Deciphering the dark proteome of Chikungunya virus. Sci Rep 2018; 8:5822. [PMID: 29643398 PMCID: PMC5895634 DOI: 10.1038/s41598-018-23969-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 03/21/2018] [Indexed: 12/24/2022] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus. The outbreak of CHIKV infection has been seen in many tropical and subtropical regions of the biosphere. Current reports evidenced that after outbreaks in 2005-06, the fitness of this virus propagating in Aedes albopictus enhanced due to the epistatic mutational changes in its envelope protein. In our study, we evaluated the prevalence of intrinsically disordered proteins (IDPs) and IDP regions (IDPRs) in CHIKV proteome. IDPs/IDPRs are known as members of a 'Dark Proteome' that defined as a set of polypeptide segments or whole protein without unique three-dimensional structure within the cellular milieu but with significant biological functions, such as cell cycle regulation, control of signaling pathways, and maintenance of viral proteomes. However, the intrinsically disordered aspects of CHIKV proteome and roles of IDPs/IDPRs in the pathogenic mechanism of this important virus have not been evaluated as of yet. There are no existing reports on the analysis of intrinsic disorder status of CHIKV. To fulfil this goal, we have analyzed the abundance and functionality of IDPs/IDPRs in CHIKV proteins, involved in the replication and maturation. It is likely that these IDPs/IDPRs can serve as novel targets for disorder based drug design.
Collapse
Affiliation(s)
- Ankur Singh
- School of Basic Sciences, Indian Institute of Technology Mandi, Himachal Pradesh, 175005, India
| | - Ankur Kumar
- School of Basic Sciences, Indian Institute of Technology Mandi, Himachal Pradesh, 175005, India
| | - Rakhi Yadav
- School of Basic Sciences, Indian Institute of Technology Mandi, Himachal Pradesh, 175005, India
| | - Vladimir N Uversky
- Department of Molecular Medicine and Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
- Laboratory of New Methods in Biology, Institute for Biological Instrumentation, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Rajanish Giri
- School of Basic Sciences, Indian Institute of Technology Mandi, Himachal Pradesh, 175005, India.
- BioX Centre, Indian Institute of Technology Mandi, VPO Kamand, 175005, India.
| |
Collapse
|
32
|
Singh A, Kumar A, Uversky V, Giri R. Understanding the interactability of chikungunya virus proteinsviamolecular recognition feature analysis. RSC Adv 2018; 8:27293-27303. [PMID: 35539973 PMCID: PMC9083250 DOI: 10.1039/c8ra04760j] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/12/2018] [Indexed: 12/27/2022] Open
Abstract
The chikungunya virus (CHIKV) is an alphavirus that has an enveloped icosahedral capsid and is transmitted byAedessp. mosquitos.
Collapse
Affiliation(s)
- Ankur Singh
- School of Basic Sciences
- Indian Institute of Technology Mandi
- Himachal Pradesh 175005
- India
| | - Ankur Kumar
- School of Basic Sciences
- Indian Institute of Technology Mandi
- Himachal Pradesh 175005
- India
| | - Vladimir N. Uversky
- Department of Molecular Medicine and Byrd Alzheimer's Research Institute
- Morsani College of Medicine
- University of South Florida
- Tampa
- USA
| | - Rajanish Giri
- School of Basic Sciences
- Indian Institute of Technology Mandi
- Himachal Pradesh 175005
- India
- BioX Centre
| |
Collapse
|
33
|
Abstract
Beginning in 2004, chikungunya virus (CHIKV) went from an endemic pathogen limited to Africa and Asia that caused periodic outbreaks to a global pathogen. Given that outbreaks caused by CHIKV have continued and expanded, serious consideration must be given to identifying potential options for vaccines and therapeutics. Currently, there are no licensed products in this realm, and control relies completely on the use of personal protective measures and integrated vector control, which are only minimally effective. Therefore, it is prudent to urgently examine further possibilities for control. Vaccines have been shown to be highly effective against vector-borne diseases. However, as CHIKV is known to rapidly spread and generate high attack rates, therapeutics would also be highly valuable. Several candidates are currently being developed; this review describes the multiple options under consideration for future development and assesses their relative advantages and disadvantages.
Collapse
|
34
|
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne arbovirus which poses a major threat to global public health. Definitive CHIKV diagnosis is crucial, especially in distinguishing the disease from dengue virus, which co-circulates in endemic areas and shares the same mosquito vectors. Laboratory diagnosis is mainly based on serological or molecular approaches. The E2 glycoprotein is a good candidate for serological diagnosis since it is the immunodominant antigen during the course of infection, and reacts with seropositive CHIKV sera. In this chapter, we describe the generation of stable clone Sf9 (Spodoptera frugiperda) cells expressing secreted, soluble, and native recombinant CHIKV E2 glycoprotein. We use direct plasmid expression in insect cells, rather than the traditional technique of generating recombinant baculovirus. This recombinant protein is useful for serological diagnosis of CHIKV infection.
Collapse
|
35
|
Ramsey J, Mukhopadhyay S. Disentangling the Frames, the State of Research on the Alphavirus 6K and TF Proteins. Viruses 2017; 9:v9080228. [PMID: 28820485 PMCID: PMC5580485 DOI: 10.3390/v9080228] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 08/03/2017] [Accepted: 08/16/2017] [Indexed: 01/04/2023] Open
Abstract
For 30 years it was thought the alphavirus 6K gene encoded a single 6 kDa protein. However, through a bioinformatics search 10 years ago, it was discovered that there is a frameshifting event and two proteins, 6K and transframe (TF), are translated from the 6K gene. Thus, many functions attributed to the 6K protein needed reevaluation to determine if they properly belong to 6K, TF, or both proteins. In this mini-review, we reevaluate the past research on 6K and put those results in context where there are two proteins, 6K and TF, instead of one. Additionally, we discuss the most cogent outstanding questions for 6K and TF research, including their collective importance in alphavirus budding and their potential importance in disease based on the latest virulence data.
Collapse
Affiliation(s)
- Jolene Ramsey
- Department of Biology at Indiana University, Bloomington, IN 47405, USA.
| | | |
Collapse
|
36
|
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus in the family Togaviridae that causes outbreaks of debilitating acute and chronic arthralgia in humans. Although historically associated with localized outbreaks in Africa and Asia, recent epidemics in the Indian Ocean region and the Americas have led to the recognition that CHIKV is capable of moving into previously unaffected areas and causing significant levels of human suffering. The severity of CHIKV rheumatic disease, which can severely impact life quality of infected individuals for weeks, months, or even years, combined with the explosive nature of CHIKV outbreaks and its demonstrated ability to quickly spread into new regions, has led to renewed interest in developing strategies for the prevention or treatment of CHIKV-induced disease. Therefore, this chapter briefly discusses the biology of CHIKV and the factors contributing to CHIKV dissemination, while also discussing the pathogenesis of CHIKV-induced disease and summarizing the status of efforts to develop safe and effective therapies and vaccines against CHIKV and related viruses.
Collapse
|
37
|
Göertz GP, Vogels CBF, Geertsema C, Koenraadt CJM, Pijlman GP. Mosquito co-infection with Zika and chikungunya virus allows simultaneous transmission without affecting vector competence of Aedes aegypti. PLoS Negl Trop Dis 2017; 11:e0005654. [PMID: 28570693 PMCID: PMC5469501 DOI: 10.1371/journal.pntd.0005654] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 06/13/2017] [Accepted: 05/19/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Zika virus (ZIKV) and chikungunya virus (CHIKV) are highly pathogenic arthropod-borne viruses that are currently a serious health burden in the Americas, and elsewhere in the world. ZIKV and CHIKV co-circulate in the same geographical regions and are mainly transmitted by Aedes aegypti mosquitoes. There is a growing number of case reports of ZIKV and CHIKV co-infections in humans, but it is uncertain whether co-infection occurs via single or multiple mosquito bites. Here we investigate the potential of Ae. aegypti mosquitoes to transmit both ZIKV and CHIKV in one bite, and we assess the consequences of co-infection on vector competence. METHODOLOGY/PRINCIPAL FINDINGS First, growth curves indicated that co-infection with CHIKV negatively affects ZIKV production in mammalian, but not in mosquito cells. Next, Ae. aegypti mosquitoes were infected with ZIKV, CHIKV, or co-infected via an infectious blood meal or intrathoracic injections. Infection and transmission rates, as well as viral titers of positive mosquitoes, were determined at 14 days after blood meal or 7 days after injection. Saliva and bodies of (co-)infected mosquitoes were scored concurrently for the presence of ZIKV and/or CHIKV using a dual-colour immunofluorescence assay. The results show that orally exposed Ae. aegypti mosquitoes are highly competent, with transmission rates of up to 73% for ZIKV, 21% for CHIKV, and 12% of mosquitoes transmitting both viruses in one bite. However, simultaneous oral exposure to both viruses did not change infection and transmission rates compared to exposure to a single virus. Intrathoracic injections indicate that the selected strain of Ae. aegypti has a strong salivary gland barrier for CHIKV, but a less profound barrier for ZIKV. CONCLUSIONS/SIGNIFICANCE This study shows that Ae. aegypti can transmit both ZIKV and CHIKV via a single bite. Furthermore, co-infection of ZIKV and CHIKV does not influence the vector competence of Ae. aegypti.
Collapse
Affiliation(s)
- Giel P. Göertz
- Laboratory of Virology, Wageningen University & Research, Wageningen, The Netherlands
| | - Chantal B. F. Vogels
- Laboratory of Entomology, Wageningen University & Research, Wageningen, The Netherlands
| | - Corinne Geertsema
- Laboratory of Virology, Wageningen University & Research, Wageningen, The Netherlands
| | | | - Gorben P. Pijlman
- Laboratory of Virology, Wageningen University & Research, Wageningen, The Netherlands
| |
Collapse
|
38
|
Thiemmeca S, Tamdet C, Punyadee N, Prommool T, Songjaeng A, Noisakran S, Puttikhunt C, Atkinson JP, Diamond MS, Ponlawat A, Avirutnan P. Secreted NS1 Protects Dengue Virus from Mannose-Binding Lectin-Mediated Neutralization. THE JOURNAL OF IMMUNOLOGY 2016; 197:4053-4065. [PMID: 27798151 DOI: 10.4049/jimmunol.1600323] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 09/16/2016] [Indexed: 12/16/2022]
Abstract
Flavivirus nonstructural protein 1 (NS1) is a unique secreted nonstructural glycoprotein. Although it is absent from the flavivirus virion, intracellular and extracellular forms of NS1 have essential roles in viral replication and the pathogenesis of infection. The fate of NS1 in insect cells has been more controversial, with some reports suggesting it is exclusively cell associated. In this study, we confirm NS1 secretion from cells of insect origin and characterize its physical, biochemical, and functional properties in the context of dengue virus (DENV) infection. Unlike mammalian cell-derived NS1, which displays both high mannose and complex type N-linked glycans, soluble NS1 secreted from DENV-infected insect cells contains only high mannose glycans. Insect cell-derived secreted NS1 also has different physical properties, including smaller and more heterogeneous sizes and the formation of less stable NS1 hexamers. Both mammalian and insect cell-derived NS1 bind to complement proteins C1s, C4, and C4-binding protein, as well as to a novel partner, mannose-binding lectin. Binding of NS1 to MBL protects DENV against mannose-binding lectin-mediated neutralization by the lectin pathway of complement activation. As we detected secreted NS1 and DENV together in the saliva of infected Aedes aegypti mosquitoes, these findings suggest a mechanism of viral immune evasion at the very earliest phase of infection.
Collapse
Affiliation(s)
- Somchai Thiemmeca
- Division of Dengue Hemorrhagic Fever Research, Department of Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.,Graduate Program, Department of Immunology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Chamaiporn Tamdet
- Division of Dengue Hemorrhagic Fever Research, Department of Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Nuntaya Punyadee
- Division of Dengue Hemorrhagic Fever Research, Department of Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Tanapan Prommool
- Medical Biotechnology Research Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Bangkok 12120, Thailand
| | - Adisak Songjaeng
- Division of Dengue Hemorrhagic Fever Research, Department of Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Sansanee Noisakran
- Division of Dengue Hemorrhagic Fever Research, Department of Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.,Medical Biotechnology Research Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Bangkok 12120, Thailand
| | - Chunya Puttikhunt
- Division of Dengue Hemorrhagic Fever Research, Department of Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.,Medical Biotechnology Research Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Bangkok 12120, Thailand
| | - John P Atkinson
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110.,Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110.,Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110; and
| | - Alongkot Ponlawat
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Panisadee Avirutnan
- Division of Dengue Hemorrhagic Fever Research, Department of Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; .,Medical Biotechnology Research Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Bangkok 12120, Thailand
| |
Collapse
|
39
|
Metz SW, Tian S, Hoekstra G, Yi X, Stone M, Horvath K, Miley MJ, DeSimone J, Luft CJ, de Silva AM. Precisely Molded Nanoparticle Displaying DENV-E Proteins Induces Robust Serotype-Specific Neutralizing Antibody Responses. PLoS Negl Trop Dis 2016; 10:e0005071. [PMID: 27764114 PMCID: PMC5072622 DOI: 10.1371/journal.pntd.0005071] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 09/24/2016] [Indexed: 12/13/2022] Open
Abstract
Dengue virus (DENV) is the causative agent of dengue fever and dengue hemorrhagic fever. The virus is endemic in over 120 countries, causing over 350 million infections per year. Dengue vaccine development is challenging because of the need to induce simultaneous protection against four antigenically distinct DENV serotypes and evidence that, under some conditions, vaccination can enhance disease due to specific immunity to the virus. While several live-attenuated tetravalent dengue virus vaccines display partial efficacy, it has been challenging to induce balanced protective immunity to all 4 serotypes. Instead of using whole-virus formulations, we are exploring the potentials for a particulate subunit vaccine, based on DENV E-protein displayed on nanoparticles that have been precisely molded using Particle Replication in Non-wetting Template (PRINT) technology. Here we describe immunization studies with a DENV2-nanoparticle vaccine candidate. The ectodomain of DENV2-E protein was expressed as a secreted recombinant protein (sRecE), purified and adsorbed to poly (lactic-co-glycolic acid) (PLGA) nanoparticles of different sizes and shape. We show that PRINT nanoparticle adsorbed sRecE without any adjuvant induces higher IgG titers and a more potent DENV2-specific neutralizing antibody response compared to the soluble sRecE protein alone. Antigen trafficking indicate that PRINT nanoparticle display of sRecE prolongs the bio-availability of the antigen in the draining lymph nodes by creating an antigen depot. Our results demonstrate that PRINT nanoparticles are a promising platform for delivering subunit vaccines against flaviviruses such as dengue and Zika.
Collapse
Affiliation(s)
- Stefan W. Metz
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Shaomin Tian
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Gabriel Hoekstra
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Xianwen Yi
- Lineberger Comprehensive Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Michelle Stone
- Liquidia Technologies, Research Triangle Park, North Carolina, United States of America
| | - Katie Horvath
- Liquidia Technologies, Research Triangle Park, North Carolina, United States of America
| | - Michael J. Miley
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Joseph DeSimone
- Lineberger Comprehensive Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, United States of America
- Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Chris J. Luft
- Lineberger Comprehensive Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Aravinda M. de Silva
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
40
|
The viral capping enzyme nsP1: a novel target for the inhibition of chikungunya virus infection. Sci Rep 2016; 6:31819. [PMID: 27545976 PMCID: PMC4992889 DOI: 10.1038/srep31819] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 07/18/2016] [Indexed: 11/26/2022] Open
Abstract
The chikungunya virus (CHIKV) has become a substantial global health threat due to its massive re-emergence, the considerable disease burden and the lack of vaccines or therapeutics. We discovered a novel class of small molecules ([1,2,3]triazolo[4,5-d]pyrimidin-7(6H)-ones) with potent in vitro activity against CHIKV isolates from different geographical regions. Drug-resistant variants were selected and these carried a P34S substitution in non-structural protein 1 (nsP1), the main enzyme involved in alphavirus RNA capping. Biochemical assays using nsP1 of the related Venezuelan equine encephalitis virus revealed that the compounds specifically inhibit the guanylylation of nsP1. This is, to the best of our knowledge, the first report demonstrating that the alphavirus capping machinery is an excellent antiviral drug target. Considering the lack of options to treat CHIKV infections, this series of compounds with their unique (alphavirus-specific) target offers promise for the development of therapy for CHIKV infections.
Collapse
|
41
|
Bala Murugan S, Sathishkumar R. Chikungunya infection: A potential re-emerging global threat. ASIAN PAC J TROP MED 2016; 9:933-937. [PMID: 27794385 DOI: 10.1016/j.apjtm.2016.07.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 06/18/2016] [Accepted: 07/17/2016] [Indexed: 10/21/2022] Open
Abstract
Infectious diseases are indeed a lifelong threat to everyone irrespective of age, sex, lifestyle and socio-economic status. The infectious diseases have persisted among the prominent causes of death globally. Recently, re-emergence of Chikungunya viral infection harmed many in Asian and African countries. Chikungunya was considered as a major threat in developing and under-developed countries; the recent epidemiological outbreak of Chikungunya in La Reunion urges the global researchers to develop effective vaccine against this viral disease. In this review, Chikungunya, pathogenesis and epidemiology were briefly described.
Collapse
Affiliation(s)
- Shanmugaraj Bala Murugan
- Plant Genetic Engineering Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India.
| | - Ramalingam Sathishkumar
- Plant Genetic Engineering Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India.
| |
Collapse
|
42
|
Ahola T, Couderc T, Courderc T, Ng LFP, Hallengärd D, Powers A, Lecuit M, Esteban M, Merits A, Roques P, Liljeström P. Therapeutics and vaccines against chikungunya virus. Vector Borne Zoonotic Dis 2016; 15:250-7. [PMID: 25897811 DOI: 10.1089/vbz.2014.1681] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Currently, there are no licensed vaccines or therapies available against chikungunya virus (CHIKV), and these were subjects discussed during a CHIKV meeting recently organized in Langkawi, Malaysia. In this review, we chart the approaches taken in both areas. Because of a sharp increase in new data in these fields, the present paper is complementary to previous reviews by Weaver et al. in 2012 and Kaur and Chu in 2013 . The most promising antivirals so far discovered are reviewed, with a special focus on the virus-encoded replication proteins as potential targets. Within the vaccines in development, our review emphasizes the various strategies in parallel development that are unique in the vaccine field against a single disease.
Collapse
Affiliation(s)
- Tero Ahola
- 1 Department of Food and Environmental Sciences, University of Helsinki , Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Volz A, Lim S, Kaserer M, Lülf A, Marr L, Jany S, Deeg CA, Pijlman GP, Koraka P, Osterhaus ADME, Martina BE, Sutter G. Immunogenicity and protective efficacy of recombinant Modified Vaccinia virus Ankara candidate vaccines delivering West Nile virus envelope antigens. Vaccine 2016; 34:1915-26. [PMID: 26939903 DOI: 10.1016/j.vaccine.2016.02.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/14/2016] [Accepted: 02/16/2016] [Indexed: 12/30/2022]
Abstract
West Nile virus (WNV) cycles between insects and wild birds, and is transmitted via mosquito vectors to horses and humans, potentially causing severe neuroinvasive disease. Modified Vaccinia virus Ankara (MVA) is an advanced viral vector for developing new recombinant vaccines against infectious diseases and cancer. Here, we generated and evaluated recombinant MVA candidate vaccines that deliver WNV envelope (E) antigens and fulfil all the requirements to proceed to clinical testing in humans. Infections of human and equine cell cultures with recombinant MVA demonstrated efficient synthesis and secretion of WNV envelope proteins in mammalian cells non-permissive for MVA replication. Prime-boost immunizations in BALB/c mice readily induced circulating serum antibodies binding to recombinant WNV E protein and neutralizing WNV in tissue culture infections. Vaccinations in HLA-A2.1-/HLA-DR1-transgenic H-2 class I-/class II-knockout mice elicited WNV E-specific CD8+ T cell responses. Moreover, the MVA-WNV candidate vaccines protected C57BL/6 mice against lineage 1 and lineage 2 WNV infection and induced heterologous neutralizing antibodies. Thus, further studies are warranted to evaluate these recombinant MVA-WNV vaccines in other preclinical models and use them as candidate vaccine in humans.
Collapse
Affiliation(s)
- Asisa Volz
- German Centre for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Veterinaerstrasse 13, D-80539 Munich, Germany
| | - Stephanie Lim
- Viroscience Lab, Erasmus Medical Center, Rotterdam, The Netherlands; Artemis One Health Research Institute, Utrecht, The Netherlands
| | - Martina Kaserer
- German Centre for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Veterinaerstrasse 13, D-80539 Munich, Germany
| | - Anna Lülf
- German Centre for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Veterinaerstrasse 13, D-80539 Munich, Germany
| | - Lisa Marr
- German Centre for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Veterinaerstrasse 13, D-80539 Munich, Germany
| | - Sylvia Jany
- German Centre for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Veterinaerstrasse 13, D-80539 Munich, Germany
| | - Cornelia A Deeg
- Institute for Animal Physiology, LMU University of Munich, Munich, Germany
| | - Gorben P Pijlman
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands
| | - Penelope Koraka
- Viroscience Lab, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Albert D M E Osterhaus
- Viroscience Lab, Erasmus Medical Center, Rotterdam, The Netherlands; Artemis One Health Research Institute, Utrecht, The Netherlands
| | - Byron E Martina
- Viroscience Lab, Erasmus Medical Center, Rotterdam, The Netherlands; Artemis One Health Research Institute, Utrecht, The Netherlands
| | - Gerd Sutter
- German Centre for Infection Research (DZIF), Institute for Infectious Diseases and Zoonoses, LMU University of Munich, Veterinaerstrasse 13, D-80539 Munich, Germany.
| |
Collapse
|
44
|
Next generation sequencing of DNA-launched Chikungunya vaccine virus. Virology 2016; 490:83-90. [PMID: 26855330 DOI: 10.1016/j.virol.2016.01.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 01/11/2016] [Accepted: 01/13/2016] [Indexed: 11/22/2022]
Abstract
Chikungunya virus (CHIKV) represents a pandemic threat with no approved vaccine available. Recently, we described a novel vaccination strategy based on iDNA® infectious clone designed to launch a live-attenuated CHIKV vaccine from plasmid DNA in vitro or in vivo. As a proof of concept, we prepared iDNA plasmid pCHIKV-7 encoding the full-length cDNA of the 181/25 vaccine. The DNA-launched CHIKV-7 virus was prepared and compared to the 181/25 virus. Illumina HiSeq2000 sequencing revealed that with the exception of the 3' untranslated region, CHIKV-7 viral RNA consistently showed a lower frequency of single-nucleotide polymorphisms than the 181/25 RNA including at the E2-12 and E2-82 residues previously identified as attenuating mutations. In the CHIKV-7, frequencies of reversions at E2-12 and E2-82 were 0.064% and 0.086%, while in the 181/25, frequencies were 0.179% and 0.133%, respectively. We conclude that the DNA-launched virus has a reduced probability of reversion mutations, thereby enhancing vaccine safety.
Collapse
|
45
|
de Pinheiro CGM, Pedrosa MDO, Teixeira NC, Ano Bom APD, van Oers MM, Oliveira GGDS. Optimization of canine interleukin-12 production using a baculovirus insect cell expression system. BMC Res Notes 2016; 9:36. [PMID: 26795376 PMCID: PMC4722752 DOI: 10.1186/s13104-016-1843-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 01/08/2016] [Indexed: 12/11/2022] Open
Abstract
Background Interleukin-12 is an important cytokine in mediating cellular immune responses. Results Recombinant single-chain canine IL-12 was produced in a baculovirus-insect cell system with the aim of conducting further studies on modulation of immune responses in dogs. To optimize the production of recombinant canine IL-12, a classical baculovirus and a modified vector (chitinase A and v-cathepsin knockout) were used containing a native or an optimized insert of canine IL-12.
The optimized IL-12 construct contained the GP64 signal peptide and was synthesized with optimized codons for expression in Trichoplusia ni cells. Dot-blot and Western blot analysis showed the highest production levels of recombinant IL-12 protein by the use of the modified baculovirus vector containing the optimized insert, at a multiplicity of infection of five and at 48 h after infection. The recombinant cytokine was successfully purified and showed a good degree of purity, integrity, folding, and yield, with very little endotoxin contamination. Recombinant canine IL-12 induced IFN-γ in canine lymphocytes, indicating that it was biologically active. Conclusion Therefore, this study describes an efficient method to produce adequate amounts of biologically active canine IL-12, useful for immunomodulation studies in dogs.
Collapse
Affiliation(s)
- Cristiane Garboggini Melo de Pinheiro
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil. .,Programa Nacional de Pós Doutorado-CAPES/Programa de Pós-graduação em Biotecnologia em Saúde e Medicina Investigativa, Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil.
| | | | | | - Ana Paula Dinis Ano Bom
- Laboratório de Macromoléculas, Bio-Manguinhos, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.
| | - Monique M van Oers
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands.
| | - Geraldo Gileno de Sá Oliveira
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil. .,Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais (INCT-DT), Salvador, Brazil.
| |
Collapse
|
46
|
Kuo SC, Teng CY, Ho YJ, Chen YJ, Wu TY. Using Bicistronic Baculovirus Expression Vector System to Screen the Compounds That Interfere with the Infection of Chikungunya Virus. Methods Mol Biol 2016; 1426:263-72. [PMID: 27233279 DOI: 10.1007/978-1-4939-3618-2_24] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Chikungunya virus (CHIKV) is the etiologic agent of Chikungunya fever and has emerged in many countries over the past decade. There are no effective drugs for controlling the disease. A bicistronic baculovirus expression system was utilized to co-express CHIKV structural proteins C (capsid), E2 and E1 and the enhanced green fluorescence protein (EGFP) in Spodoptera frugiperda insect cells (Sf21). The EGFP-positive Sf21 cells fused with each other and with uninfected cells to form a syncytium is mediated by the CHIKV E1 allowing it to identify chemicals that can prevent syncytium formation. The compounds characterized by this method could be anti-CHIKV drugs.
Collapse
Affiliation(s)
- Szu-Cheng Kuo
- Institute of Prevention Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Chao-Yi Teng
- Bioengineering Group, Institute of Biologics, Development Center for Biotechnology, New Taipei, Taiwan
| | - Yi-Jung Ho
- Institute of Prevention Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Ying-Ju Chen
- Bioengineering Group, Institute of Biologics, Development Center for Biotechnology, New Taipei, Taiwan
| | - Tzong-Yuan Wu
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li, Taiwan.
| |
Collapse
|
47
|
Metz SW, Pijlman GP. Production of Chikungunya Virus-Like Particles and Subunit Vaccines in Insect Cells. Methods Mol Biol 2016; 1426:297-309. [PMID: 27233282 DOI: 10.1007/978-1-4939-3618-2_27] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Chikungunya virus is a reemerging human pathogen that causes debilitating arthritic disease in humans. Like dengue and Zika virus, CHIKV is transmitted by Aedes mosquitoes in an epidemic urban cycle, and is now rapidly spreading through the Americas since its introduction in the Caribbean in late 2013. There are no licensed vaccines or antiviral drugs available, and only a few vaccine candidates have passed Phase I human clinical trials. Using recombinant baculovirus expression technology, we have generated CHIKV glycoprotein subunit and virus-like particle (VLP) vaccines that are amenable to large scale production in insect cells. These vaccines, in particular the VLPs, have shown high immunogenicity and protection against CHIKV infection in different animal models of CHIKV-induced disease. Here, we describe the production, purification, and characterization of these potent CHIKV vaccine candidates.
Collapse
Affiliation(s)
- Stefan W Metz
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708 NW, Wageningen, The Netherlands
| | - Gorben P Pijlman
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708 NW, Wageningen, The Netherlands.
| |
Collapse
|
48
|
Rahpeyma M, Fotouhi F, Makvandi M, Ghadiri A, Samarbaf-Zadeh A. Crimean-Congo Hemorrhagic Fever Virus Gn Bioinformatic Analysis and Construction of a Recombinant Bacmid in Order to Express Gn by Baculovirus Expression System. Jundishapur J Microbiol 2015; 8:e25502. [PMID: 26862379 PMCID: PMC4740762 DOI: 10.5812/jjm.25502] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 06/28/2015] [Accepted: 08/04/2015] [Indexed: 11/23/2022] Open
Abstract
Background Crimean-Congo hemorrhagic fever virus (CCHFV) is a member of the nairovirus, a genus in the Bunyaviridae family, which causes a life threatening disease in human. Currently, there is no vaccine against CCHFV and detailed structural analysis of CCHFV proteins remains undefined. The CCHFV M RNA segment encodes two viral surface glycoproteins known as Gn and Gc. Viral glycoproteins can be considered as key targets for vaccine development. Objectives The current study aimed to investigate structural bioinformatics of CCHFV Gn protein and design a construct to make a recombinant bacmid to express by baculovirus system. Materials and Methods To express the Gn protein in insect cells that can be used as antigen in animal model vaccine studies. Bioinformatic analysis of CCHFV Gn protein was performed and designed a construct and cloned into pFastBacHTb vector and a recombinant Gn-bacmid was generated by Bac to Bac system. Results Primary, secondary, and 3D structure of CCHFV Gn were obtained and PCR reaction with M13 forward and reverse primers confirmed the generation of recombinant bacmid DNA harboring Gn coding region under polyhedron promoter. Conclusions Characterization of the detailed structure of CCHFV Gn by bioinformatics software provides the basis for development of new experiments and construction of a recombinant bacmid harboring CCHFV Gn, which is valuable for designing a recombinant vaccine against deadly pathogens like CCHFV.
Collapse
Affiliation(s)
- Mehdi Rahpeyma
- Health Research Institute, Infectious and Tropical Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fatemeh Fotouhi
- Influenza Research Laboratory, Department of Virology, Pasteur Institute of Iran, Tehran, IR Iran
| | - Manouchehr Makvandi
- Department of Virology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran
| | - Ata Ghadiri
- Cellular and Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran
| | - Alireza Samarbaf-Zadeh
- Health Research Institute, Infectious and Tropical Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Corresponding author: Alireza Samarbaf-Zadeh, Health Research Institute, Infectious and Tropical Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. Tel/Fax: +98-6113738313, E-mail:
| |
Collapse
|
49
|
Hikke MC, Geertsema C, Wu V, Metz SW, van Lent JW, Vlak JM, Pijlman GP. Alphavirus capsid proteins self-assemble into core-like particles in insect cells: A promising platform for nanoparticle vaccine development. Biotechnol J 2015; 11:266-73. [PMID: 26287127 DOI: 10.1002/biot.201500147] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 06/18/2015] [Accepted: 08/13/2015] [Indexed: 11/07/2022]
Abstract
The mosquito-borne chikungunya virus (CHIKV) causes arthritic diseases in humans, whereas the aquatic salmonid alphavirus (SAV) is associated with high mortality in aquaculture of salmon and trout. Using modern biotechnological approaches, promising vaccine candidates based upon highly immunogenic, enveloped virus-like particles (eVLPs) have been developed. However, the eVLP structure (core, lipid membrane, surface glycoproteins) is more complex than that of non-enveloped, protein-only VLPs, which are structurally and morphologically 'simple'. In order to develop an alternative to alphavirus eVLPs, in this paper we engineered recombinant baculovirus vectors to produce high levels of alphavirus core-like particles (CLPs) in insect cells by expression of the CHIKV and SAV capsid proteins. The CLPs localize in dense nuclear bodies within the infected cell nucleus and are purified through a rapid and scalable protocol involving cell lysis, sonication and low-speed centrifugation steps. Furthermore, an immunogenic epitope from the alphavirus E2 glycoprotein can be successfully fused to the N-terminus of the capsid protein without disrupting the CLP self-assembling properties. We propose that immunogenic epitope-tagged alphavirus CLPs produced in insect cells present a simple and perhaps more stable alternative to alphavirus eVLPs.
Collapse
Affiliation(s)
- Mia C Hikke
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands
| | - Corinne Geertsema
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands
| | - Vincen Wu
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands
| | - Stefan W Metz
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands
| | - Jan W van Lent
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands
| | - Just M Vlak
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands
| | - Gorben P Pijlman
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands.
| |
Collapse
|
50
|
Fros JJ, Geertsema C, Zouache K, Baggen J, Domeradzka N, van Leeuwen DM, Flipse J, Vlak JM, Failloux AB, Pijlman GP. Mosquito Rasputin interacts with chikungunya virus nsP3 and determines the infection rate in Aedes albopictus. Parasit Vectors 2015; 8:464. [PMID: 26384002 PMCID: PMC4573678 DOI: 10.1186/s13071-015-1070-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/03/2015] [Indexed: 01/22/2023] Open
Abstract
Background Chikungunya virus (CHIKV) is an arthritogenic alphavirus (family Togaviridae), transmitted by Aedes species mosquitoes. CHIKV re-emerged in 2004 with multiple outbreaks worldwide and recently reached the Americas where it has infected over a million individuals in a rapidly expanding epidemic. While alphavirus replication is well understood in general, the specific function (s) of non-structural protein nsP3 remain elusive. CHIKV nsP3 modulates the mammalian stress response by preventing stress granule formation through sequestration of G3BP. In mosquitoes, nsP3 is a determinant of vector specificity, but its functional interaction with mosquito proteins is unclear. Methods In this research we studied the domains required for localization of CHIKV nsP3 in insect cells and demonstrated its molecular interaction with Rasputin (Rin), the mosquito homologue of G3BP. The biological involvement of Rin in CHIKV infection was investigated in live Ae. albopictus mosquitoes. Results In insect cells, nsP3 localized as cytoplasmic granules, which was dependent on the central domain and the C-terminal variable region but independent of the N-terminal macrodomain. Ae. albopictus Rin displayed a diffuse, cytoplasmic localization, but was effectively sequestered into nsP3-granules upon nsP3 co-expression. Site-directed mutagenesis showed that the Rin-nsP3 interaction involved the NTF2-like domain of Rin and two conserved TFGD repeats in the C-terminal variable domain of nsP3. Although in vitro silencing of Rin did not impact nsP3 localization or CHIKV replication in cell culture, Rin depletion in vivo significantly decreased the CHIKV infection rate and transmissibility in Ae.albopictus. Conclusions We identified the nsP3 hypervariable C-terminal domain as a critical factor for granular localization and sequestration of mosquito Rin. Our study offers novel insight into a conserved virus-mosquito interaction at the molecular level, and reveals a strong proviral role for G3BP homologue Rin in live mosquitoes, making the nsP3-Rin interaction a putative target to interfere with the CHIKV transmission cycle.
Collapse
Affiliation(s)
- Jelke J Fros
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708 PB, Wageningen, The Netherlands.
| | - Corinne Geertsema
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708 PB, Wageningen, The Netherlands.
| | - Karima Zouache
- Institut Pasteur, Department of Virology, Arboviruses and Insect Vectors, 25-28 rue du Docteur Roux, 75724, Paris, cedex 15, France.
| | - Jim Baggen
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708 PB, Wageningen, The Netherlands.
| | - Natalia Domeradzka
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708 PB, Wageningen, The Netherlands.
| | - Daniël M van Leeuwen
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708 PB, Wageningen, The Netherlands.
| | - Jacky Flipse
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708 PB, Wageningen, The Netherlands.
| | - Just M Vlak
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708 PB, Wageningen, The Netherlands.
| | - Anna-Bella Failloux
- Institut Pasteur, Department of Virology, Arboviruses and Insect Vectors, 25-28 rue du Docteur Roux, 75724, Paris, cedex 15, France.
| | - Gorben P Pijlman
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708 PB, Wageningen, The Netherlands.
| |
Collapse
|