1
|
Karim SU, Nazneen F, Denyoh PMD, Bai DS, Romero DG, Bai F. Heterozygous interferon signaling deficient mice as animal models for Chikungunya virus infection in the heart. Sci Rep 2025; 15:18022. [PMID: 40410198 PMCID: PMC12102211 DOI: 10.1038/s41598-025-02191-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 05/12/2025] [Indexed: 05/25/2025] Open
Abstract
Although chikungunya virus (CHIKV)-caused cardiovascular diseases are frequently reported in clinics, the underlying mechanisms are poorly understood, which is primarily due to a lack of animal models. In this study, we report that CHIKV infection in homozygous interferon α/β receptor-deficient (ifnar1-/-) and interferon α/β/γ receptor-deficient (ifnag-/-) mice resulted in high viral loads in the hearts as early as day (D) 1 post-infection (p.i.) but with 100% mortality within three days p.i. In contrast, the heterozygous ifnar1+/-and ifnag+/- mice survived CHIKV infection and bore higher viral burdens in the heart tissues than the wild-type (WT) controls. Immunohistochemistry and flow cytometry revealed that more leukocytes, particularly neutrophils, infiltrated the heart of ifnag+/- and ifnar1+/- mice than WT mice. In addition, the Hematoxylin and Eosin staining analysis showed that CHIKV infection caused vasculitis in the left ventricles on D5 p.i. in both heterozygous groups and the vacuole formation and pyknosis in ifnar1+/- mice. Moreover, CHIKV infection may also lead to cardiac fibrosis, as indicated by the upregulation of the expression of the Connective Tissue Growth Factor gene in the hearts of ifnar1+/- mice. In summary, our data suggest that the heterozygous ifnar1+/- and ifnag+/- mice are invaluable for studying pathogenesis and testing therapeutic interventions for CHIKV-caused cardiac diseases.
Collapse
Affiliation(s)
- Shazeed-Ul Karim
- Cell and Molecular Biology Program, School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, 118 College Drive # 5018, Hattiesburg, MS, 39406, USA
| | - Farzana Nazneen
- Cell and Molecular Biology Program, School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, 118 College Drive # 5018, Hattiesburg, MS, 39406, USA
| | - Prince M D Denyoh
- Cell and Molecular Biology Program, School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, 118 College Drive # 5018, Hattiesburg, MS, 39406, USA
| | - David S Bai
- Oak Grove High School, Hattiesburg, MS, 39402, USA
| | - Damian G Romero
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
- Mississippi Center of Excellence in Perinatal Research, Jackson, MS, 39216, USA
- Women's Health Research Center, Jackson, MS, 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Fengwei Bai
- Cell and Molecular Biology Program, School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, 118 College Drive # 5018, Hattiesburg, MS, 39406, USA.
| |
Collapse
|
2
|
Ma B, Cao Z, Ding W, Zhang X, Xiang Y, Cao D. Structural basis for the recognition of two different types of receptors by Western equine encephalitis virus. Cell Rep 2025; 44:115724. [PMID: 40402741 DOI: 10.1016/j.celrep.2025.115724] [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: 01/15/2025] [Revised: 03/25/2025] [Accepted: 04/29/2025] [Indexed: 05/24/2025] Open
Abstract
Western equine encephalitis virus (WEEV) enters cells via various receptors. Here, we report the cryoelectron microscopy (cryo-EM) structures of WEEV in complex with its receptors PCDH10 and very-low-density lipoprotein receptor (VLDLR). Structural analysis shows that PCDH10 binds in the cleft formed by adjacent E2-E1 heterodimers of WEEV through its EC1 ectodomain. Residues of viral envelope proteins involved in the interactions with PCDH10 EC1 are unique to WEEV. The strain-specific receptor VLDLR binds WEEV strain McMillan through two consecutive ecto-LDLR class A (LA) repeats. LA1-2, LA2-3, LA3-4, LA4-5, and LA5-6 of VLDLR all have detectable interactions with WEEV. Detailed structures of WEEV in complex with LA1-2 and LA2-3 show that the N-terminal LA repeat binds in the cleft and that the C-terminal LA repeat is attached to the E2 B domain. The acquisition of a single E2 mutation (V265F) allows WEEV strain 71V-1658, originally unable to bind VLDLR, to gain this receptor-binding ability. The binding of VLDLR to WEEV is in a mode different from those of other alphaviruses.
Collapse
Affiliation(s)
- Bingting Ma
- Beijing Frontier Research Center for Biological Structure, Center for Infectious Disease Research, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, P.R. China; SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan 030001, P.R. China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, P.R. China
| | - Ziyi Cao
- National Laboratory of Biomacromolecules, Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences (CAS), Beijing 100101, P.R. China; University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Weijia Ding
- Beijing Frontier Research Center for Biological Structure, Center for Infectious Disease Research, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, P.R. China; SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan 030001, P.R. China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, P.R. China
| | - Xinzheng Zhang
- National Laboratory of Biomacromolecules, Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences (CAS), Beijing 100101, P.R. China; University of Chinese Academy of Sciences, Beijing 100049, P.R. China.
| | - Ye Xiang
- Beijing Frontier Research Center for Biological Structure, Center for Infectious Disease Research, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, P.R. China; SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan 030001, P.R. China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, P.R. China.
| | - Duanfang Cao
- National Laboratory of Biomacromolecules, Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences (CAS), Beijing 100101, P.R. China; University of Chinese Academy of Sciences, Beijing 100049, P.R. China.
| |
Collapse
|
3
|
Fritzer A, Suhrbier A, Hugo LE, Tang B, Devine G, Jost S, Meinke AL. Assessment of the transmission of live-attenuated chikungunya virus vaccine VLA1553 by Aedes albopictus mosquitoes. Parasit Vectors 2025; 18:171. [PMID: 40355954 PMCID: PMC12070702 DOI: 10.1186/s13071-025-06789-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 04/01/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Chikungunya virus (CHIKV) is a mosquito-transmitted, arthritogenic alphavirus that causes sporadic outbreaks of often debilitating rheumatic disease. The recently approved CHIKV vaccine, IXCHIQ, is based on a live-attenuated CHIKV strain (VLA1553), with viraemic vaccine recipients theoretically able to transmit VLA1553 to mosquitoes with ensuing onward transmission. We thus evaluated VLA1553 transmission from artificial blood meals to Aedes albopictus mosquitoes, and onward transmission to mice. METHODS Female A. albopictus mosquitoes were fed on defibrinated sheep blood containing wild-type CHIKV (viral titre: 7.50 log10CCID50/mL) or VLA1553 (viral titres: 7.85, 5.72, 4.58, and 3.79 log10CCID50/mL). Viral titres in mosquito bodies and saliva were determined using CCID50 assays 7-8 days after the blood meal. After providing CHIKV or VLA1553 (viral titres ~ 7-8 log10CCID50/mL) in blood meals to mosquitoes, infected mosquitoes were fed on highly susceptible Irf3/7-/- mice (n = 3 per group). Data were re-analysed using the same reverse transcription quantitative polymerase chain reaction (RT-qPCR) as for an earlier VLA1553 phase 1 clinical trial, to allow correlations between blood meal titres and viraemia in vaccine recipients. RESULTS Mosquito body viral titres were significantly higher (P < 0.0001) for CHIKV versus VLA1553-fed mosquitoes at blood meal viral titres of ~ 7-8 log10CCID50/mL. Mosquito body VLA1553 titres decreased with reducing blood meal titres, but there was no dose-dependent effect on saliva viral titres. No dissemination to salivary glands was seen at blood meal titres ≤ 3.875 log10CCID50/mL. CHIKV-fed mosquitoes were able to transmit virus, and induce viraemia in, 3/3 Irf3/7-/- mice via mosquito bites. In contrast, 0/3 Irf3/7-/- mice became infected after bites from VLA1553-fed mosquitoes. RT-qPCR comparisons with phase 1 clinical data for VLA1553-vaccinated individuals indicated that VLA1553 viraemia was at or below the aforementioned threshold for transmission. CONCLUSIONS The evidence presented herein argue that the low viraemia in VLA1553-vaccinated individuals would mitigate against transmission. In addition, replication of VLA1553 in mosquito bodies was also significantly attenuated. Overall, mosquito-borne transmission of VLA1553 from vaccinated individuals to others appears improbable.
Collapse
Affiliation(s)
- Andrea Fritzer
- Valneva Austria GmbH, Campus Vienna Biocenter 3, 1030, Vienna, Austria.
| | - Andreas Suhrbier
- Inflammation Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4029, Australia
| | - Leon E Hugo
- Mosquito Control, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4029, Australia
| | - Bing Tang
- Inflammation Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4029, Australia
| | - Greg Devine
- Mosquito Control, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4029, Australia
| | - Sandra Jost
- Valneva Austria GmbH, Campus Vienna Biocenter 3, 1030, Vienna, Austria
| | - Andreas L Meinke
- Valneva Austria GmbH, Campus Vienna Biocenter 3, 1030, Vienna, Austria
| |
Collapse
|
4
|
Ou TP, Sorn S, Nguon K, In S, Ken S, Ly S, Flamand C, Voirin N, Mandron M, Watson H, Duong V. Viral Kinetics During Acute Chikungunya Virus Infection: Insights Into Potential Role of Monoclonal Antibodies in Viral Clearance and Prophylaxis Using Mathematical Modeling. J Med Virol 2025; 97:e70391. [PMID: 40358000 DOI: 10.1002/jmv.70391] [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: 01/23/2025] [Revised: 03/31/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025]
Abstract
Chikungunya virus (CHIKV), an arthritogenic alphavirus, is a significant public health threat in endemic and newly affected regions. This study investigates viral kinetics, immune responses, and the potential of monoclonal antibody (mAb) therapies to mitigate viraemia and transmission during acute CHIKV infection, providing novel insights into early intervention strategies. Using data from 29 patients in Cambodia, serial sampling and viral load quantification revealed that the population-average peak viral load occurred ~1.87 days prior to symptom onset. Children demonstrated higher peak viral loads and faster replication rates compared to adults, although symptom severity and burden were similar across age groups. IgM antibodies appeared earlier in adults (median: 4.1 days) than in children (median: 5.1 days; p = 0.036). C-reactive protein (CRP) levels were transiently elevated in about 50% of patients but showed no correlation with disease severity. Mathematical modeling highlighted that prophylactic mAb therapies, when administered 3 days before symptoms onset, could substantially reduce viral load and potentially prevent detectable viraemia. While these findings underscore the potential of mAbs as an early therapeutic strategy, further studies are necessary to evaluate the robustness of these results and assess their practical implications to curb CHIKV outbreaks by minimizing viraemia and presymptomatic transmission.
Collapse
Affiliation(s)
- Tey Putita Ou
- Virology Unit, Pasteur Network, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Sopheak Sorn
- Epidemiology and Public Health Unit, Pasteur Network, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Kunthy Nguon
- Epidemiology and Public Health Unit, Pasteur Network, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Saraden In
- Virology Unit, Pasteur Network, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Sreymom Ken
- Virology Unit, Pasteur Network, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Sowath Ly
- Epidemiology and Public Health Unit, Pasteur Network, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Claude Flamand
- Epidemiology and Public Health Unit, Pasteur Network, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
- Mathematical Modelling of Infectious Diseases Unit, Institut Pasteur, Université Paris Cité, U1332 INSERM, UMR2000 CNRS, Paris, France
| | | | - Marie Mandron
- Clinical Development and Translational Medicine, Evotec ID, Lyon, France
| | - Hugh Watson
- Clinical Development and Translational Medicine, Evotec ID, Lyon, France
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Veasna Duong
- Virology Unit, Pasteur Network, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| |
Collapse
|
5
|
Suzuki Y. Application of reverse genetics system to Chikungunya virus study. Virology 2025; 605:110465. [PMID: 40043635 DOI: 10.1016/j.virol.2025.110465] [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: 12/06/2024] [Revised: 02/21/2025] [Accepted: 02/22/2025] [Indexed: 03/16/2025]
Abstract
Chikungunya virus (CHIKV) is an enveloped RNA virus of the Togaviridae family that causes Chikungunya fever, characterized by fever, myalgia, and arthralgia. Although the mortality rate attributed to CHIKV infection is low, the risk of severe disease increases in young children, the elderly, and people with medical conditions. Given the significant impact of these clinical manifestations, an effective regimen for the treatment of CHIKV infection is needed. The reverse genetics system, an approach to generate a complete virus from cloned cDNA, has been widely used to characterize the replication and pathogenicity of medically important viruses. In particular, the implementation of reverse genetics allows researchers to manipulate the viral genome in vitro, contributing to the development of vaccines and antivirals. This review will present the status of the application of the reverse genetics system to advance knowledge of the biological aspects of CHIKV and summarize how this technology is being used to establish preventive and therapeutic measures against CHIKV infection.
Collapse
Affiliation(s)
- Youichi Suzuki
- Department of Microbiology and Infection Control, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan.
| |
Collapse
|
6
|
Matsumura R, Bannai H, Nemoto M, Higa Y, Kai I, Sasaki T, Futami K, Yoshikawa A, Fujita R, Hino M, Nagata K, Kuwata R, Kaku Y, Kobayashi D, Minakawa N, Kasai S, Itoyama K, Maeda K, Isawa H. Genetic, Phylogenetic, and Serological Analysis of a Getah Virus Strain Isolated from Culex tritaeniorhynchus Mosquitoes in Nagasaki, Japan in 2022. Jpn J Infect Dis 2025; 78:71-78. [PMID: 39617482 DOI: 10.7883/yoken.jjid.2024.250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Getah virus (GETV), belonging to the genus Alphavirus within the family Togaviridae, is a mosquito-borne virus that causes fever, rash, edema in horses, fatalities, and pregnancy disorders in pigs. It has caused occasional outbreaks in horse populations in Japan, China, and India, and the endemic areas are gradually expanding, particularly in Asia and Oceania. In this study, we isolated a new GETV strain from Culex tritaeniorhynchus mosquitoes collected in Nagasaki Prefecture, Japan, in 2022. Phylogenetic analysis demonstrated that this new strain, 22IH8, was more closely related to previous Chinese strains than to the strains prevalent in Japan in the 2010s. Furthermore, some amino acid substitutions in the viral proteins of strain 22IH8 were similar to those in previous Chinese strains. These results indicate that strain 22IH8 may have recently invaded the Japanese archipelago from mainland Asia. The antiserum against the current vaccine strain MI-110 demonstrated high neutralization activity against the strain 22IH8, indicating the efficiency of the current vaccine for horses in Japan.
Collapse
Affiliation(s)
- Ryo Matsumura
- Department of Medical Entomology, National Institute of Infectious Diseases, Japan
- Graduate School of Agriculture, Meiji University, Japan
| | - Hiroshi Bannai
- Equine Research Institute, Japan Racing Association, Japan
| | - Manabu Nemoto
- Equine Research Institute, Japan Racing Association, Japan
| | - Yukiko Higa
- Department of Medical Entomology, National Institute of Infectious Diseases, Japan
| | - Izumi Kai
- Department of Medical Entomology, National Institute of Infectious Diseases, Japan
- Graduate School of Agriculture, Meiji University, Japan
| | - Toshinori Sasaki
- Department of Medical Entomology, National Institute of Infectious Diseases, Japan
| | - Kyoko Futami
- Department of Vector Ecology and Environment, Institute of Tropical Medicine, Nagasaki University, Japan
| | - Akira Yoshikawa
- Nagasaki Prefectural Institute for Environmental Research and Public Health, Japan
| | | | - Masato Hino
- Faculty of Agriculture, Kyushu University, Japan
| | - Kosuke Nagata
- Faculty of Agriculture, Kyushu University, Japan
- Hokkaido Research Organization, Japan
| | - Ryusei Kuwata
- Faculty of Veterinary Medicine, Okayama University of Science, Japan
| | - Yoshihiro Kaku
- Department of Veterinary Science, National Institute of Infectious Diseases, Japan
| | - Daisuke Kobayashi
- Department of Medical Entomology, National Institute of Infectious Diseases, Japan
- Management Department of Biosafety, Laboratory Animal, and Pathogen Bank, National Institute of Infectious Diseases, Japan
| | - Noboru Minakawa
- Department of Vector Ecology and Environment, Institute of Tropical Medicine, Nagasaki University, Japan
| | - Shinji Kasai
- Department of Medical Entomology, National Institute of Infectious Diseases, Japan
| | - Kyo Itoyama
- Graduate School of Agriculture, Meiji University, Japan
| | - Ken Maeda
- Department of Veterinary Science, National Institute of Infectious Diseases, Japan
| | - Haruhiko Isawa
- Department of Medical Entomology, National Institute of Infectious Diseases, Japan
| |
Collapse
|
7
|
Hall RA, Nguyen W, Khromykh AA, Suhrbier A. Insect-specific virus platforms for arbovirus vaccine development. Front Immunol 2025; 16:1521104. [PMID: 40160816 PMCID: PMC11949993 DOI: 10.3389/fimmu.2025.1521104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Certain insect-specific viruses (ISVs), specifically the mosquito alphaviruses, Eilat and Yada Yada viruses, and orthoflaviviruses, Binjari, Aripo, YN15-283-02 and Chaoyang viruses, have emerged as potential platforms for generation of whole virus vaccines for human and veterinary applications. These ISVs are remarkably tolerant of the substitution of their structural polyproteins with those of alphaviruses and orthoflaviviruses that are pathogenic in humans and/or animals. The resulting ISV-based chimeric vaccines have been evaluated in mouse models and have demonstrated safety and efficacy in non-human primates, crocodiles and pigs. Targets include chikungunya, Venezuelan and eastern equine encephalitis, dengue, Zika, yellow fever, Japanese encephalitis and West Nile viruses. ISV-based chimeric vaccines provide authentically folded tertiary and quaternary whole virion particle structures to the immune system, a key feature for induction of protective antibody responses. These vaccines are manufactured in C6/36 or C7-10 mosquito cell lines, where they grow to high titers, but they do not replicate in vertebrate vaccine recipients. This review discusses the progress of these emerging technologies and addresses challenges related to adjuvanting, safety, and manufacturing.
Collapse
Affiliation(s)
- Roy A. Hall
- School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, QLD, Australia
- Global Virus Network Centre of Excellence, Australian Infectious Diseases Research Centre, Brisbane, QLD, Australia
| | - Wilson Nguyen
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Alexander A. Khromykh
- School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, QLD, Australia
- Global Virus Network Centre of Excellence, Australian Infectious Diseases Research Centre, Brisbane, QLD, Australia
| | - Andreas Suhrbier
- Global Virus Network Centre of Excellence, Australian Infectious Diseases Research Centre, Brisbane, QLD, Australia
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
8
|
Duven M, Friedrichs A, Tomlinson MG, Steffen I, Gerold G. Tetraspanins 10 and 15 support Venezuelan equine encephalitis virus replication in astrocytoma cells. Mol Biol Cell 2025; 36:ar35. [PMID: 39878649 PMCID: PMC11974957 DOI: 10.1091/mbc.e24-12-0574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 01/31/2025] Open
Abstract
Tetraspanins (Tspans) are transmembrane proteins that coordinate life cycle steps of viruses from distinct families. Here, we identify the human Tspan10 and Tspan15, both members of the TspanC8 subfamily, as replication factors for alphavirus Venezuelan equine encephalitis virus (VEEV) in astrocytoma cells. Pharmacological inhibition and small interfering RNA (siRNA)-mediated silencing of TspanC8 interactor a disintegrin and metalloproteinase 10 (ADAM10) reduced VEEV infection. Silencing of Tspan10, Tspan15, and ADAM10 did not affect VEEV entry but diminished viral genome replication. We report that Tspan10 is important for VEEV infection of several cell lines, while silencing of Tspan15 diminishes infection with several alphaviruses, but not flaviviruses, in astrocytoma cells. Conversely, we demonstrate that siRNA-mediated silencing of Tspan14, another member of the TspanC8 family, enhances infection with lentiviral pseudoparticles harbouring the envelope proteins of VEEV, identifying it as a restriction factor for VEEV entry. Silencing of ADAM10/Tspan15 substrate neuronal (N)-cadherin reduced VEEV infectivity, suggesting potential roles of ADAM10 substrates in VEEV infection. In sum, our study identifies three TspanC8s and ADAM10 as important modulators of VEEV infectivity.
Collapse
Affiliation(s)
- Mara Duven
- Institute for Biochemistry and Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Alina Friedrichs
- Institute for Biochemistry and Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Michael G. Tomlinson
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, B15 2TT United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), The Universities of Birmingham and Nottingham, The Midlands, United Kingdom
| | - Imke Steffen
- Institute for Biochemistry and Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Gisa Gerold
- Institute for Biochemistry and Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, 90187 Umeå, Sweden
- Department of Clinical Microbiology, Virology, Umeå University, 90187 Umeå, Sweden
- Institute of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
9
|
Ghoshal A, Tse EG, Hossain MA, Asressu KH, Merten EM, Sears JD, Howell S, Perveen S, Burdick J, Morales NL, Martinez SA, Law I, Davenport BJ, Morrison TE, Streblow ZJ, Streblow DN, Mordant AL, Webb TS, Cabrera A, Herring LE, Arrowsmith CH, Pearce KH, Moorman NJ, Heise MT, Couñago RM, Brown PJ, Willson TM. A covalent chemical probe for Chikungunya nsP2 cysteine protease with antialphaviral activity and proteome-wide selectivity. Sci Rep 2025; 15:7264. [PMID: 40025188 PMCID: PMC11873239 DOI: 10.1038/s41598-025-91673-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/21/2025] [Indexed: 03/04/2025] Open
Abstract
Chikungunya is a mosquito-borne viral disease that causes fever and severe joint pain for which there is no direct acting drug treatments. Vinyl sulfone SGC-NSP2PRO-1 (3) was identified as a potent inhibitor of the nsP2 cysteine protease (nsP2pro) that reduced viral titer against infectious isolates of Chikungunya and other alphaviruses. The covalent warhead in 3 captured the active site C478 and inactivated nsP2pro with a kinact/Ki ratio of 5950 M-1 s-1. The vinyl sulfone 3 was inactive across a panel of 23 other cysteine proteases and demonstrated remarkable proteome-wide selectivity by two chemoproteomic methods. A negative control analog SGC-NSP2PRO-1N (4) retained the isoxazole core and covalent warhead but demonstrated > 100-fold decrease in enzyme inhibition. Both 3 and 4 were stable across a wide range of pH in solution and upon prolonged storage as solids. Vinyl sulfone 3 and its negative control 4 will find utility as high-quality chemical probes to study the role of the nsP2pro in cellular studies of alphaviral replication and virulence.
Collapse
Affiliation(s)
- Anirban Ghoshal
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Edwin G Tse
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Mohammad Anwar Hossain
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Kesatebrhan Haile Asressu
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Eric M Merten
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - John D Sears
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Stefanie Howell
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Sumera Perveen
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Structural Genomics Consortium, University of Toronto, Toronto, ON, M5G 1L7, Canada
| | - Jane Burdick
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Noah L Morales
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Sabian A Martinez
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Isabella Law
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Bennett J Davenport
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Thomas E Morrison
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Zachary J Streblow
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Daniel N Streblow
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Angie L Mordant
- UNC Metabolomics and Proteomics Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Thomas S Webb
- UNC Metabolomics and Proteomics Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Aurora Cabrera
- UNC Metabolomics and Proteomics Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Laura E Herring
- UNC Metabolomics and Proteomics Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Cheryl H Arrowsmith
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Structural Genomics Consortium, University of Toronto, Toronto, ON, M5G 1L7, Canada
| | - Kenneth H Pearce
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Nathaniel J Moorman
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Mark T Heise
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Rafael M Couñago
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Center of Medicinal Chemistry, Center for Molecular Biology and Genetic Engineering, University of Campinas, Campinas, SP, 13083-886, Brazil
| | - Peter J Brown
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Timothy M Willson
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- READDI Avidd Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
10
|
Wang S, Mahalingam S, Merits A. Alphavirus nsP2: A Multifunctional Regulator of Viral Replication and Promising Target for Anti-Alphavirus Therapies. Rev Med Virol 2025; 35:e70030. [PMID: 40064592 PMCID: PMC11893376 DOI: 10.1002/rmv.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 02/19/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
Alphaviruses are re-emerging vector-born pathogens that cause arthralgia or encephalitic diseases on a global scale. While a vaccine against chikungunya virus was recently approved, no vaccines currently exist for other alphaviruses, nor are there antiviral drugs for the treatment of alphavirus infections. Alphaviruses have positive-strand RNA genomes, and their RNA replication is coordinated by activities of the multifunctional nonstructural protein 2 (nsP2), a helicase-protease and a subunit of viral RNA replicase. We provide a comprehensive overview of nsP2 functions and inhibitors of its activities for their potential as effective antivirals. Furthermore, analysis of nsP2 activities suggests that it could be targeted to develop advanced live attenuated vaccines and strategies for controlling alphavirus transmission by mosquito vectors.
Collapse
Affiliation(s)
- Sainan Wang
- Institute of BioengineeringUniversity of TartuTartuEstonia
| | - Suresh Mahalingam
- Institute for Biomedicine and GlycomicsGriffith UniversityGold CoastAustralia
- Global Virus Network (GVN) Centre of Excellence in ArbovirusesGriffith UniversityGold CoastAustralia
- School of Pharmacy and Medical SciencesGriffith UniversityGold CoastAustralia
| | - Andres Merits
- Institute of BioengineeringUniversity of TartuTartuEstonia
| |
Collapse
|
11
|
Parashar B, Malviya R, Sridhar SB, Wadhwa T, Talath S, Shareef J. Eastern equine encephalitis virus: Pathogenesis, immune response, and clinical manifestations. INFECTIOUS MEDICINE 2025; 4:100167. [PMID: 40026316 PMCID: PMC11869868 DOI: 10.1016/j.imj.2025.100167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/16/2024] [Accepted: 01/14/2025] [Indexed: 03/05/2025]
Abstract
Eastern equine encephalitis virus (EEEV) is a lethal Alphavirus transmitted by Culiseta melanura mosquitoes that primarily cycles between birds. Although rare, infections in humans and horses are associated with high mortality rates and severe neurological effects. Climate change appears to be increasing the spread of this virus. This study aims to provide a comprehensive analysis of EEEV, including its transmission dynamics, pathogenesis, induced host immune response, and long-term impacts on survivors. It also highlights the virus's unique immune evasion strategies that complicate disease management and contribute to severe clinical outcomes, such as encephalitis with fever, convulsions, and coma. Survivors often face chronic cognitive, motor, and psychosocial impairments. Despite these significant public health risks, gaps remain in understanding the molecular mechanisms underlying immune evasion and the long-term neurological sequelae in survivors. By collating current knowledge, this review underscores the urgent need for the development of targeted vaccines and therapeutic interventions to mitigate the growing threat of EEEV, particularly in the context of climate change-driven geographical expansion.
Collapse
Affiliation(s)
- Bhumika Parashar
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 201310, Uttar Pradesh, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 201310, Uttar Pradesh, India
- Galgotias Multi-Disciplinary Research & Development Cell (G-MRDC), Galgotias University, Greater Noida 201308, Uttar Pradesh, India
| | - Sathvik Belagodu Sridhar
- RAK College of Pharmacy, RAK Medical & Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Tarun Wadhwa
- RAK College of Pharmacy, RAK Medical & Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Sirajunisa Talath
- RAK College of Pharmacy, RAK Medical & Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Javedh Shareef
- RAK College of Pharmacy, RAK Medical & Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| |
Collapse
|
12
|
Lantz AM, Baxter VK. Neuropathogenesis of Old World Alphaviruses: Considerations for the Development of Medical Countermeasures. Viruses 2025; 17:261. [PMID: 40007016 PMCID: PMC11860675 DOI: 10.3390/v17020261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Chikungunya virus (CHIKV) and other alphaviruses that primarily induce arthritogenic disease in humans, known as "Old World" alphaviruses, present an emerging public health concern as geographic ranges of mosquito vectors expand due to climate change. While a vaccine against CHIKV has recently been approved by several countries in North America and Europe, access to effective preventative countermeasures against disease induced by Old World alphaviruses remains elusive for the most vulnerable populations. Furthermore, treatment options continue to be limited to supportive care. Atypical neurological disease manifestations caused by Old World alphaviruses, which make up as many as 25% of the cases in some CHIKV outbreaks, present special challenges when considering strategies for developing effective countermeasures. This review focuses on Old World alphaviruses, specifically CHIKV, Ross River virus, O'nyoug-nyoug virus, and Mayaro virus, concentrating on the atypical neurological disease manifestations they may cause. Our current understanding of Old World alphavirus neuropathogenesis, gained from human cases and preclinical animal models, is discussed, including viral and host factors' roles in disease development. The current state of alphavirus preventatives and treatments, both virus-targeting and host-directed therapies, is then summarized and discussed in the context of addressing neurological disease induced by Old World alphaviruses.
Collapse
|
13
|
Dunagan MM, Dábilla N, McNinch C, Brenchley JM, Dolan PT, Fox JM. Interaction of the endogenous antibody response with activating FcγRs enhance control of Mayaro virus through monocytes. PLoS Pathog 2025; 21:e1012944. [PMID: 39993025 PMCID: PMC11884725 DOI: 10.1371/journal.ppat.1012944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 03/06/2025] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
Mayaro virus (MAYV) is an emerging arbovirus. Previous studies have shown antibody Fc effector functions are critical for optimal monoclonal antibody-mediated protection against alphaviruses; however, the requirement of Fc gamma receptors (FcγRs) for protection during natural infection has not been evaluated. Here, we showed mice lacking activating FcγRs (FcRγ-/-) developed prolonged clinical disease with increased MAYV in joint-associated tissues. Viral reduction was associated with anti-MAYV cell surface binding antibodies rather than neutralizing antibodies. Lack of Fc-FcγR engagement increased the number of monocytes present in the joint-associated tissue through chronic timepoints. Single-cell RNA sequencing showed elevated levels of pro-inflammatory monocytes in joint-associated tissue with increased MAYV RNA present in FcRγ-/- monocytes and macrophages. Transfer of FcRγ-/- monocytes into wild type animals was sufficient to increase virus in joint-associated tissue. Overall, this study suggests that engagement of antibody Fc with activating FcγRs promotes protective responses during MAYV infection and prevents a pro-viral role for monocytes.
Collapse
Affiliation(s)
- Megan M. Dunagan
- Emerging Virus Immunity Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Nathânia Dábilla
- Quantitative Virology and Evolution Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Colton McNinch
- Bioinformatics and Computational Bioscience Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Jason M. Brenchley
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Patrick T. Dolan
- Quantitative Virology and Evolution Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Julie M. Fox
- Emerging Virus Immunity Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
14
|
Garzan A, Ahmed SK, Haese NN, Sulgey G, Medica S, Smith JL, Zhang S, Ahmad F, Karyakarte S, Rasmussen L, DeFilippis V, Tekwani B, Bostwick R, Suto MJ, Hirsch AJ, Morrison TE, Heise MT, Augelli-Szafran CE, Streblow DN, Pathak AK, Moukha-Chafiq O. 4-Substituted-2-Thiazole Amides as Viral Replication Inhibitors of Alphaviruses. J Med Chem 2024; 67:20858-20878. [PMID: 39621435 DOI: 10.1021/acs.jmedchem.4c01073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
2-(Methylthio)-N-(4-(naphthalen-2-yl)thiazol-2-yl)nicotinamide 1 was identified as an inhibitor against Chikungunya virus (CHIKV) with good antiviral activity [EC50 = 0.6 μM; EC90 = 0.93 μM and viral titer reduction (VTR) of 6.9 logs at 10 μM concentration] with no observed cytotoxicity (CC50 = 132 μM) in normal human dermal fibroblast (NHDF) cells. Structure-activity relationship (SAR) studies to further improve the potency, efficacy, and drug-like properties of 1 led to the discovery of a new potent inhibitor N-(4-(3-((4-cyanophenyl)amino)phenyl)thiazol-2-yl)-2-(methylthio)nicotinamide 26, which showed a VTR of 8.7 logs at 10 μM against CHIKV and an EC90 of 0.45 μM with considerably improved MLM stability (t1/2 = 74 min) as compared to 1. Mechanism of action studies show that 26 inhibits alphavirus replication by blocking subgenomic viral RNA translation and structural protein synthesis. The in vivo efficacy studies of compound 26 on CHIKV infection in mice are reported.
Collapse
Affiliation(s)
- Atefeh Garzan
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - S Kaleem Ahmed
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Nicole N Haese
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, United States
| | - Gauthami Sulgey
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, United States
| | - Samuel Medica
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, United States
| | - Jessica L Smith
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, United States
| | - Sixue Zhang
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Fahim Ahmad
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Shuklendu Karyakarte
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Lynn Rasmussen
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Victor DeFilippis
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, United States
| | - Babu Tekwani
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Robert Bostwick
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Mark J Suto
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Alec J Hirsch
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, United States
| | - Thomas E Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine, 12800 E. 19th Avenue, Aurora, Colorado 80045, United States
| | - Mark T Heise
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Corinne E Augelli-Szafran
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, United States
| | - Ashish K Pathak
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Omar Moukha-Chafiq
- Scientific Platforms Division, Southern Research, 2000 ninth Avenue South, Birmingham, Alabama 35205, United States
| |
Collapse
|
15
|
Maurer G, Buerger V, Larcher-Senn J, Erlsbacher F, Dubischar K, Eder-Lingelbach S, Jaramillo JC. Pooled safety evaluation for a new single-shot live-attenuated chikungunya vaccine†. J Travel Med 2024; 31:taae133. [PMID: 39400050 DOI: 10.1093/jtm/taae133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/10/2024] [Accepted: 10/11/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Chikungunya disease, caused by chikungunya virus (CHIKV), is associated with substantial morbidity, including debilitating CHIKV-related arthralgia. METHODS Three clinical trials of a CHIKV vaccine (VLA1553, IXCHIQ®) were conducted in the USA: a Phase 1 dose-finding trial, a pivotal Phase 3 trial and a Phase 3 lot-to-lot consistency trial. Participants were healthy adults (≥18 years) and received a single intramuscular dose of VLA1553 (3520 participants) or placebo (1033 participants). Solicited injection site and systemic adverse events (AEs) (10-14 days post-vaccination), unsolicited AEs (28 and 180 days post-vaccination), AEs of special interest (AESIs) (28 days post-vaccination), medically attended AEs (MAAEs), serious AEs (SAEs) (180 days post-vaccination) and pregnancies were evaluated. Safety data were pooled, and analyses were descriptive. RESULTS Overall, 63.7% of participants receiving VLA1553 experienced AEs (44.7% for placebo) that were generally mild. Solicited injection-site AEs, solicited systemic AEs and unsolicited (Day 29) AEs were reported by 15.5, 50.9 and 22.7% of participants who received VLA1553 and 11.1, 26.9 and 13.4% who received placebo. Arthralgia was reported by 16.7% of participants who received VLA1553 and 4.8% of participants who received placebo; none required medical attention. MAAEs, AESIs and SAEs were reported by 12.4, 0.3 and 1.5% of participants who received VLA1553 and 11.3, 0.1 and 0.8% of participants who received placebo. Protocol-defined AESIs were mild and short-lived, and two VLA1553-related SAEs resolved without sequelae. There were no clinically important differences in AE incidence based on age or medical history and no VLA1553-related adverse pregnancy outcomes. There were three deaths (two in the VLA1553 group and one in the placebo group); none was vaccine-related. CONCLUSIONS A single dose of VLA1553 presented with an excellent local tolerability profile and overall safety in line with that expected for a live-attenuated vaccine. The safety profile was comparable in participants aged 18-64 years and ≥65 years.
Collapse
Affiliation(s)
- Gabriele Maurer
- Valneva Austria GmbH, Campus Vienna Biocenter 3, 1030 Vienna, Austria
| | - Vera Buerger
- Valneva Austria GmbH, Campus Vienna Biocenter 3, 1030 Vienna, Austria
| | - Julian Larcher-Senn
- Assign Data Management and Biostatistics GmbH, Stadlweg 23, 6020 Innsbruck, Austria
| | - Florian Erlsbacher
- Assign Data Management and Biostatistics GmbH, Stadlweg 23, 6020 Innsbruck, Austria
| | - Katrin Dubischar
- Valneva Austria GmbH, Campus Vienna Biocenter 3, 1030 Vienna, Austria
| | | | | |
Collapse
|
16
|
Tong K, Hernandez EM, Basore K, Fremont DH, Lai JR. Chikungunya virus E2 B domain nanoparticle immunogen elicits homotypic neutralizing antibody in mice. Vaccine 2024; 42:126405. [PMID: 39413488 PMCID: PMC11645211 DOI: 10.1016/j.vaccine.2024.126405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/18/2024]
Abstract
Alphaviruses are enveloped, positive-sense single-stranded RNA viruses that cause severe human and animal illness. Arthritogenic alphaviruses, such as Chikungunya virus (CHIKV) and Mayaro virus (MAYV), are globally distributed, transmitted by mosquitoes, and can cause rheumatic disease characterized by fever, rash, myalgia, and peripheral polyarthralgia that can persist for years post-infection. These infections can also result in more severe clinical manifestations such as hemorrhage, encephalopathy, and mortality. Several potent monoclonal antibodies (mAbs) with broad neutralizing activity have been shown to bind to the E2 B domain (E2-B) of the alphavirus glycoprotein, suggesting that E2-B epitopes are a site of susceptibility for multiple arthritogenic alphaviruses. However, it is unknown whether E2-B alone can elicit a broadly neutralizing humoral response. Here, we generate and characterize nanoparticle-based immunogens containing CHIKV and MAYV E2-B. Immunization with the CHIKV E2-B nanoparticle elicited sera that were cross-reactive toward CHIKV and MAYV E2-B, but had only homotypic neutralizing activity (serum titer of 1:512) against CHIKV vaccine strain 181/25. Furthermore, immunization with MAYV E2-B nanoparticles elicited non-neutralizing antibody, but sera were cross-reactive for both CHIKV and MAYV E2-B. Our findings suggest that the immunodominant epitopes within CHIKV and MAYV E2-B are bound by cross-reactive, but not cross-neutralizing antibody. Therefore, development of broad E2-B based vaccines that induce broadly neutralizing antibody responses will require engineering to alter the immunodominant landscape.
Collapse
Affiliation(s)
- Karen Tong
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Erica M Hernandez
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Katherine Basore
- Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Daved H Fremont
- Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jonathan R Lai
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
17
|
McMahon R, Toepfer S, Sattler N, Schneider M, Narciso-Abraham M, Hadl S, Hochreiter R, Kosulin K, Mader R, Zoihsl O, Wressnigg N, Dubischar K, Buerger V, Eder-Lingelbach S, Jaramillo JC. Antibody persistence and safety of a live-attenuated chikungunya virus vaccine up to 2 years after single-dose administration in adults in the USA: a single-arm, multicentre, phase 3b study. THE LANCET. INFECTIOUS DISEASES 2024; 24:1383-1392. [PMID: 39146946 DOI: 10.1016/s1473-3099(24)00357-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/14/2024] [Accepted: 05/24/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Chikungunya virus infection can lead to long-term debilitating symptoms. A precursor phase 3 clinical study showed high seroprotection (defined as a 50% plaque reduction of chikungunya virus-specific neutralising antibodies on a micro plaque reduction neutralisation test [μPRNT] titre of ≥150 in baseline seronegative participants) up to 6 months after a single vaccination of the chikungunya virus vaccine VLA1553 (Valneva Austria, Vienna, Austria) and a good safety profile. Here we report antibody persistence and safety up to 2 years. METHODS In this single-arm, multicentre, phase 3b study, we recruited participants from the precursor phase 3 trial from professional vaccine trial sites in the USA. Participants (aged ≥18 years) were eligible if they had completed the previous study and received VLA1553. Chikungunya virus-specific neutralising antibodies were evaluated at 28 days, 6 months, and 1 year and 2 years after vaccination. The primary outcome was the proportion of seroprotected participants (ie, μPRNT50 titre of ≥150) at 1 and 2 years, assessed in all eligible participants who had at least one post-vaccination immunogenicity sample available, overall and by age group at the time of vaccination (18-64 years and ≥65 years). Adverse events of special interest at the time of transition from the previous study to the current study (ie, at 6 months) and serious adverse events during the current study were recorded (ie, between 6 months and 2 years). All analyses were descriptive. This study is registered with ClinicalTrials.gov, NCT04838444, and immunogenicity follow-up is ongoing. FINDINGS In the precursor study, participants were screened between Sept 17, 2020, and April 10, 2021; data cutoff for this analysis was March 31, 2023. Of 2724 participants in the precursor study who received one dose of VLA1553, 363 participants were analysed in this study (310 [85%] aged 18-64 years and 53 [15%] aged ≥65 years at enrolment in the precursor study; mean age 47·7 years [SD 14·2], 207 [57%] of 363 participants were female, 156 [43%] were male, 280 [77%] were White, and 314 [87%] were not Hispanic or Latino). Strong seroprotection was observed at 1 year (98·9% [356 of 360 assessable participants; 97·2-99·7]) and 2 years (96·8% [306 of 316; 94·3-98·5]) after vaccination, and was very similar between those aged 18-64 years (at 1 year: 98·7% [303 of 307; 96·7-99·6]; at 2 years: 96·6% [256 of 265; 93·7-98·4]) and those aged 65 years and older (at 1 year: 100% [53 of 53; 93·3-100]; at 2 years: 98·0% [50 of 51; 89·6-100]) at each timepoint. No adverse events of special interest were ongoing at the time of transition. Ten serious adverse events occurred in nine (2%) participants between the 6-month and 2-year timepoints, including one death (due to drug overdose) that was determined to not be related to VLA1553. INTERPRETATION After a single VLA1553 vaccination, chikungunya virus-neutralising antibodies above the threshold considered to be protective persisted up to 2 years and there were no long-term serious adverse events related to vaccination. VLA1553 is an efficient and safe intervention that offers high seroprotection against chikungunya virus infection, a virus likely to spread globally with an urgent demand for long-lasting prophylaxis. FUNDING Valneva Austria, Coalition for Epidemic Preparedness Innovation, and EU Horizon 2020.
Collapse
|
18
|
Allen SW, Ribeiro Dos Santos G, Paul KK, Paul R, Rahman MZ, Alam MS, Rahman M, Al-Amin HM, Vanhomwegen J, Weaver SC, Smull T, Lee KH, Gurley ES, Salje H. Results of a Nationally Representative Seroprevalence Survey of Chikungunya Virus in Bangladesh. J Infect Dis 2024; 230:e1031-e1038. [PMID: 38942731 PMCID: PMC11565896 DOI: 10.1093/infdis/jiae335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/12/2024] [Accepted: 06/26/2024] [Indexed: 06/30/2024] Open
Abstract
There is an increasing global burden from chikungunya virus (CHIKV). Bangladesh reported a major epidemic in 2017, but it was unclear whether there had been prior widespread transmission. We conducted a nationally representative seroprevalence survey in 70 randomly selected communities immediately before the epidemic. We found that 69 of 2938 sampled individuals (2.4%) were seropositive to CHIKV. Seropositivity to dengue virus (adjusted odds ratio, 3.13 [95% confidence interval, 1.86-5.27]), male sex (0.59 [.36-.99]), and community presence of Aedes aegypti mosquitoes (1.80 [1.05-3.0]7) were significantly associated with CHIKV seropositivity. Using a spatial prediction model, we estimated that across the country, 4.99 (95% confidence interval, 4.89-5.08) million people had been previously infected. These findings highlight high population susceptibility before the major outbreak and that previous outbreaks must have been spatially isolated.
Collapse
Affiliation(s)
- Sam W Allen
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | | | - Kishor K Paul
- Kirby Institute, University of New South Wales, Sydney, Australia
- School of Population Health, University of New South Wales, Sydney, New South Wales, Australia
- One Health Laboratory, icddr,b, Dhaka, Bangladesh
| | - Repon Paul
- One Health Laboratory, icddr,b, Dhaka, Bangladesh
- Centre for Big Data Research in Health, University of New South Wales, Sydney, New South Wales, Australia
| | | | | | | | - Hasan Mohammad Al-Amin
- One Health Laboratory, icddr,b, Dhaka, Bangladesh
- QIMR Berghofer Medical Research Institute, The University of Queensland, Herston, Australia
- School of the Environment, The University of Queensland, Herston, Queensland, Australia
| | - Jessica Vanhomwegen
- Institut Pasteur, Université Paris Cité, Unité Environnement et Risques Infectieux, Cellule d'Intervention Biologique d'Urgence (CIBU), 75015 Paris, France
| | - Scott C Weaver
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Taylor Smull
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Maryland, Baltimore, USA
| | - Kyu Han Lee
- Emory Global Health Institute, Emory University, Atlanta, Georgia, USA
| | - Emily S Gurley
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Maryland, Baltimore, USA
| | - Henrik Salje
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
19
|
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
|
20
|
Fritsch H, Giovanetti M, Clemente LG, da Rocha Fernandes G, Fonseca V, de Lima MM, Falcão M, de Jesus N, de Cerqueira EM, Venâncio da Cunha R, de Oliveira Francisco MVL, de Siqueira IC, de Oliveira C, Xavier J, Ferreira JGG, Queiroz FR, Smith E, Tisoncik-Go J, Van Voorhis WC, Rabinowitz PM, Wasserheit JN, Gale M, de Filippis AMB, Alcantara LCJ. Unraveling the Complexity of Chikungunya Virus Infection Immunological and Genetic Insights in Acute and Chronic Patients. Genes (Basel) 2024; 15:1365. [PMID: 39596565 PMCID: PMC11593632 DOI: 10.3390/genes15111365] [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: 10/01/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
Background: The chikungunya virus (CHIKV), transmitted by infected Aedes mosquitoes, has caused a significant number of infections worldwide. In Brazil, the emergence of the CHIKV-ECSA genotype in 2014 posed a major public health challenge due to its association with more severe symptoms. Objectives/Methods: This study aimed to shed new light on the host immune response by examining the whole-blood transcriptomic profile of both CHIKV-acute and chronically infected individuals from Feira de Santana, Bahia, Brazil, a region heavily affected by CHIKV, Dengue, and Zika virus epidemics. Results: Our data reveal complex symptomatology characterized by arthralgia and post-chikungunya neuropathy in individuals with chronic sequelae, particularly affecting women living in socially vulnerable situations. Analysis of gene modules suggests heightened metabolic processes, represented by an increase in NADH, COX5A, COA3, CYC1, and cap methylation in patients with acute disease. In contrast, individuals with chronic manifestations exhibit a distinct pattern of histone methylation, probably mediated by NCOA3 in the coactivation of different nuclear receptors, KMT2 genes, KDM3B and TET2, and with alterations in the immunological response, majorly led by IL-17RA, IL-6R, and STAT3 Th17 genes. Conclusion: Our results emphasize the complexity of CHIKV disease progression, demonstrating the heterogeneous gene expression and symptomatologic scenario across both acute and chronic phases. Moreover, the identification of specific gene modules associated with viral pathogenesis provides critical insights into the molecular mechanisms underlying these distinct clinical manifestations.
Collapse
Affiliation(s)
- Hegger Fritsch
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (H.F.); (J.X.)
- Institut National de la Santé et de la Recherche Médicale, U1259—MAVIVHe, Université de Tours, 37032 Tours, France
| | - Marta Giovanetti
- Department of Science and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, 00128 Rome, Italy;
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-002, Brazil;
- Climate Amplified Diseases and Epidemics (CLIMADE)—CLIMADE Americas, Belo Horizonte 30190-002, Brazil
| | - Luan Gaspar Clemente
- Escola Superior de Agricultura Luiz de Queiroz, Departamento de Zootecnia, Universidade de São Paulo, Piracicaba 13418-900, Brazil;
| | | | - Vagner Fonseca
- Departamento de Ciências Exatas e da Terra, Universidade Estadual da Bahia, Salvador 41150-000, Brazil;
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Maricelia Maia de Lima
- Departamento de Saúde, Universidade Estadual de Feira de Santana, Feira de Santana 44036-900, Brazil; (M.M.d.L.); (E.M.d.C.)
- Secretaria de Municipal de Saúde de Feira de Santana, Divisão de Vigilância Epidemiológica, Feira de Santana 44027-010, Brazil; (M.F.); (N.d.J.)
| | - Melissa Falcão
- Secretaria de Municipal de Saúde de Feira de Santana, Divisão de Vigilância Epidemiológica, Feira de Santana 44027-010, Brazil; (M.F.); (N.d.J.)
| | - Neuza de Jesus
- Secretaria de Municipal de Saúde de Feira de Santana, Divisão de Vigilância Epidemiológica, Feira de Santana 44027-010, Brazil; (M.F.); (N.d.J.)
| | - Erenilde Marques de Cerqueira
- Departamento de Saúde, Universidade Estadual de Feira de Santana, Feira de Santana 44036-900, Brazil; (M.M.d.L.); (E.M.d.C.)
| | | | | | | | - Carla de Oliveira
- Laboratório de Arbovírus e Vírus Hemorrágicos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil;
| | - Joilson Xavier
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (H.F.); (J.X.)
| | - Jorge Gomes Goulart Ferreira
- Núcleo de Ensino, Pesquisa e Inovação, Instituto Mário Penna, Belo Horizonte 30380-420, Brazil; (J.G.G.F.); (F.R.Q.)
| | - Fábio Ribeiro Queiroz
- Núcleo de Ensino, Pesquisa e Inovação, Instituto Mário Penna, Belo Horizonte 30380-420, Brazil; (J.G.G.F.); (F.R.Q.)
| | - Elise Smith
- Department of Immunology, University of Washington, Seattle, WA 98109, USA; (E.S.); (J.T.-G.); (M.G.J.)
| | - Jennifer Tisoncik-Go
- Department of Immunology, University of Washington, Seattle, WA 98109, USA; (E.S.); (J.T.-G.); (M.G.J.)
| | | | - Peter M. Rabinowitz
- Departments of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA;
| | | | - Michael Gale
- Department of Immunology, University of Washington, Seattle, WA 98109, USA; (E.S.); (J.T.-G.); (M.G.J.)
| | - Ana Maria Bispo de Filippis
- Laboratório de Arbovírus e Vírus Hemorrágicos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil;
| | - Luiz Carlos Junior Alcantara
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-002, Brazil;
- Climate Amplified Diseases and Epidemics (CLIMADE)—CLIMADE Americas, Belo Horizonte 30190-002, Brazil
| |
Collapse
|
21
|
Buerger V, Maurer G, Kosulin K, Hochreiter R, Larcher-Senn J, Dubischar K, Eder-Lingelbach S. Combined immunogenicity evaluation for a new single-dose live-attenuated chikungunya vaccine. J Travel Med 2024; 31:taae084. [PMID: 38959854 DOI: 10.1093/jtm/taae084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/07/2024] [Accepted: 06/12/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Chikungunya is a serious and debilitating viral infection with a significant disease burden. VLA1553 (IXCHIQ®) is a live-attenuated vaccine licensed for active immunization for prevention of disease caused by chikungunya virus (CHIKV). METHODS Immunogenicity following a single dose of VLA1553 was evaluated in healthy adults aged ≥18 years in two Phase 3 trials [N = 656 participants (per protocol analysis set)]. Immunogenicity data to 180 days post-vaccination [geometric mean titres (GMTs), seroresponse rate, seroconversion rate] were pooled for the two trials. A comparison of subgroups based on age, sex, body mass index (BMI), race and baseline seropositivity was included. All analyses were descriptive. RESULTS Most participants were aged 18-64 years (N = 569/656 [86.7%]), there were slightly more females (N = 372/656 [56.7%]), most were not Hispanic/Latino (N = 579/656 [88.3%]), and most were White (N = 517/656 [78.8%]). In baseline seronegative participants, GMT peaked at Day 29 post-vaccination, and subsequently declined slightly but remained elevated until Day 180. At Days 29, 85 and 180, seroresponse rate was 98.3, 97.7 and 96.4% and seroconversion rate was 98.5, 98.4 and 98.2%. There were no differences in seroresponse rate in participants aged 18-64 years or ≥65 years at Day 29 (98.1 vs 100%), Day 85 (97.4 vs 100%) and Day 180 (96.3 vs 96.5%) nor based on sex, BMI, ethnicity or race. An immune response was shown in a small heterogenous population of baseline seropositive participants, with GMTs showing the same trend as baseline seronegative participants. CONCLUSIONS A single dose of VLA1553 elicited a very strong immune response by Day 29 that remained elevated at Day 180 in both baseline seronegative and seropositive participants in a combined evaluation of two Phase 3 trials. The vaccine was similarly immunogenic in participants aged ≥65 years and 18-64 years, and there were no differences based on subgroup analyses for sex, BMI, ethnicity or race.
Collapse
Affiliation(s)
- Vera Buerger
- Valneva Austria GmbH, Campus Vienna Biocenter, 3103 Vienna, Austria
| | - Gabriele Maurer
- Valneva Austria GmbH, Campus Vienna Biocenter, 3103 Vienna, Austria
| | - Karin Kosulin
- Valneva Austria GmbH, Campus Vienna Biocenter, 3103 Vienna, Austria
| | | | - Julian Larcher-Senn
- Assign Data Management and Statistics GmbH, Stadlweg 23, 6020 Innsbruck, Austria
| | - Katrin Dubischar
- Valneva Austria GmbH, Campus Vienna Biocenter, 3103 Vienna, Austria
| | | |
Collapse
|
22
|
Chen LH, Fritzer A, Hochreiter R, Dubischar K, Meyer S. From bench to clinic: the development of VLA1553/IXCHIQ, a live-attenuated chikungunya vaccine. J Travel Med 2024; 31:taae123. [PMID: 39255380 PMCID: PMC11497415 DOI: 10.1093/jtm/taae123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/14/2024] [Accepted: 09/09/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND Over the past 20 years, over 5 million cases of chikungunya, a mosquito-transmitted viral disease, have been reported in over 110 countries. Until recently, preventative strategies for chikungunya were largely ineffective, relying on vector control and individual avoidance of mosquito bites. METHODS This review outlines the preclinical and clinical efficacy and safety data that led to the approval of VLA1553 (IXCHIQ®), a live-attenuated vaccine against chikungunya disease. It also describes the innovative development pathway of VLA1553, based on an immunological surrogate of protection, and discusses ongoing and future post-licensure studies. RESULTS In mice and non-human primate models, VLA1553 elicited high titres of neutralizing antibodies, conferred protection against wild-type chikungunya virus challenge and raised no safety concerns. A Phase 1 clinical trial of VLA1553 demonstrated 100% seroconversion among 120 healthy participants, with sustained neutralizing antibody titres after 12 months. These results and determination of a surrogate marker of protection led to advancement of VLA1553 directly into Phase 3 clinical development, as agreed with the US Food and Drug Administration (FDA) and the European Medicines Agency. The pivotal Phase 3 trial met its primary immunogenicity endpoint, achieving seroprotective levels based on immuno-bridging in baseline seronegative participants 28 days post-vaccination. These findings enabled submission of a Biologics Licence Application to the FDA for accelerated approval of VLA1553 in the US for adults aged ≥18 years. Ongoing and planned studies will confirm the clinical efficacy/effectiveness and safety of VLA1553 in adults and younger individuals, and will generate data in chikungunya endemic countries that have the highest unmet need. CONCLUSION VLA1553 is the first vaccine approved for the prevention of chikungunya disease in adults, following accelerated development based on a serological surrogate marker of protection. VLA1553 adds to strategies to reduce the spread and burden of chikungunya in endemic populations and travellers.
Collapse
Affiliation(s)
- Lin H Chen
- Department of Medicine, Division of Infectious Diseases and Travel Medicine, Mount Auburn Hospital, 330 Mt Auburn St, Cambridge, MA 02138, USA
- Faculty of Medicine, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA
| | - Andrea Fritzer
- Pre-Clinical Vaccine Development Department, Valneva Austria GmbH, Campus-Vienna-Biocenter 3, 1030 Vienna, Austria
| | - Romana Hochreiter
- Clinical Serology Department, Valneva Austria GmbH, Campus-Vienna-Biocenter 3, 1030 Vienna, Austria
| | - Katrin Dubischar
- R&D Management, Valneva Austria GmbH, Campus-Vienna-Biocenter 3, 1030 Vienna, Austria
| | - Stéphanie Meyer
- Corporate Medical Affairs, Valneva SE, Ilot Saint-Joseph Bureaux Convergence, 12 ter Quai Perrache Bâtiment A, 69002 Lyon, France
| |
Collapse
|
23
|
Hameed M, Daamen AR, Hossain MS, Coutermarsh-Ott S, Lipsky PE, Weger-Lucarelli J. Obesity-Associated Changes in Immune Cell Dynamics During Alphavirus Infection Revealed by Single Cell Transcriptomic Analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.10.617696. [PMID: 39416014 PMCID: PMC11482886 DOI: 10.1101/2024.10.10.617696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Obesity induces diverse changes in host immunity, resulting in worse disease outcomes following infection with various pathogens, including arthritogenic alphaviruses. However, the impact of obesity on the functional landscape of immune cells during arthritogenic alphavirus infection remains unexplored. Here, we used single-cell RNA sequencing (scRNA-seq) to dissect the blood and tissue immune responses to Mayaro virus (MAYV) infection in lean and obese mice. Footpad injection of MAYV caused significant shifts in immune cell populations and induced robust expression of interferon response and proinflammatory cytokine genes and related pathways in both blood and tissue. In MAYV-infected lean mice, analysis of the local tissue response revealed a unique macrophage subset with high expression of IFN response genes that was not found in obese mice. This was associated with less severe inflammation in lean mice. These results provide evidence for a unique macrophage population that may contribute to the superior capacity of lean mice to control arthritogenic alphavirus infection.
Collapse
Affiliation(s)
- Muddassar Hameed
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- Department of Pathology & Immunology, Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrea R. Daamen
- AMPEL BioSolutions LLC and the RILITE Research Institute, Charlottesville, VA, United States
| | - Md Shakhawat Hossain
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Sheryl Coutermarsh-Ott
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
| | - Peter E. Lipsky
- AMPEL BioSolutions LLC and the RILITE Research Institute, Charlottesville, VA, United States
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| |
Collapse
|
24
|
Ghoshal A, Asressu KH, Hossain MA, Brown PJ, Nandakumar M, Vala A, Merten EM, Sears JD, Law I, Burdick JE, Morales NL, Perveen S, Pearce KH, Popov KI, Moorman NJ, Heise MT, Willson TM. Structure Activity of β-Amidomethyl Vinyl Sulfones as Covalent Inhibitors of Chikungunya nsP2 Cysteine Protease with Antialphavirus Activity. J Med Chem 2024; 67:16505-16532. [PMID: 39235978 PMCID: PMC11440497 DOI: 10.1021/acs.jmedchem.4c01346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024]
Abstract
Despite their widespread impact on human health, there are no approved drugs for combating alphavirus infections. The heterocyclic β-aminomethyl vinyl sulfone RA-0002034 (1a) is a potent irreversible covalent inhibitor of the alphavirus nsP2 cysteine protease with broad-spectrum antiviral activity. Analogs of 1a that varied each of the three regions of the molecule were synthesized to establish structure-activity relationships for the inhibition of Chikungunya (CHIKV) nsP2 protease and viral replication. The vinyl sulfone covalent warhead was highly sensitive to modifications. However, alterations to the core five-membered heterocycle and aryl substituent were well tolerated. The 5-(2,5-dimethoxyphenyl)pyrazole (1o) and 4-cyanopyrazole (8d) analogs exhibited kinact/Ki ratios >9000 M-1 s-1. 3-Arylisoxazole (10) was identified as an isosteric replacement for the five-membered heterocycle, which circumvented the intramolecular cyclization of pyrazole-based inhibitors like 1a. A ligand-based model of the enzyme active site was developed to aid the design of nsP2 protease inhibitors as potential therapeutics against alphaviruses.
Collapse
Affiliation(s)
- Anirban Ghoshal
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- READDI
AViDD Center, University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
| | - Kesatebrhan Haile Asressu
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- READDI
AViDD Center, University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
| | - Mohammad Anwar Hossain
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- READDI
AViDD Center, University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
| | - Peter J. Brown
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- READDI
AViDD Center, University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
| | - Meganathan Nandakumar
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Anand Vala
- Piramal
Discovery Solutions, Pharmaceutical Special Economic Zone, Sarkhej, Bavla Highway, Ahmedabad, Gujarat 382213, India
| | - Eric M. Merten
- READDI
AViDD Center, University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
- UNC
Eshelman School of Pharmacy, Center for Integrative Chemical Biology
and Drug Discovery, University of North
Carolina at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
| | - John D. Sears
- READDI
AViDD Center, University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
- Department
of Microbiology and Immunology, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Isabella Law
- Department
of Genetics, University of North Carolina
at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Jane E. Burdick
- Department
of Genetics, University of North Carolina
at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Noah L. Morales
- Department
of Genetics, University of North Carolina
at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Sumera Perveen
- Structural
Genomics Consortium, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Kenneth H. Pearce
- READDI
AViDD Center, University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
- UNC
Eshelman School of Pharmacy, Center for Integrative Chemical Biology
and Drug Discovery, University of North
Carolina at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
| | - Konstantin I. Popov
- READDI
AViDD Center, University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
- UNC
Eshelman School of Pharmacy, Center for Integrative Chemical Biology
and Drug Discovery, University of North
Carolina at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
| | - Nathaniel J. Moorman
- READDI
AViDD Center, University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
- Department
of Microbiology and Immunology, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Mark T. Heise
- READDI
AViDD Center, University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
- Department
of Microbiology and Immunology, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department
of Genetics, University of North Carolina
at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Timothy M. Willson
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- READDI
AViDD Center, University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
| |
Collapse
|
25
|
Clipes MVS, Vicente CR, Cardoso da Silva TC, Resende LC, Cerutti Junior C. Analysis of spatial and demographic factors associated with chikungunya in Espírito Santo state, Brazil. Trans R Soc Trop Med Hyg 2024; 118:597-604. [PMID: 38650504 DOI: 10.1093/trstmh/trae019] [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: 10/16/2023] [Revised: 01/08/2024] [Accepted: 03/20/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Chikungunya (CHIK) emerged in Brazil in 2014 and since then several epidemics have been observed. This study aims to describe the spatial, social and demographic characteristics of individuals affected by CHIK in Espírito Santo state. METHODS A cross-sectional study was performed using data from individuals with a confirmed diagnosis of CHIK in Espírito Santo state, Brazil, from 2018 to 2020. Monthly incidence was calculated and annual spatial distribution maps were constructed. Statistical analysis using the χ2 test identified associations between disease occurrence and sociodemographic variables. RESULTS In the period and area analysed, a CHIK epidemic occurred in 2020, with an incidence of 219.8 cases per 100 000 inhabitants. The southern and central regions of Espirito Santo state harboured a risk five times greater than the others in the epidemic region. Females (odds ratio [OR] 1.65 [95% confidence interval {CI} 1.58 to 1.72]), black people (OR 1.22 [95% CI 1.13 to 1.33]), individuals with ≤11 y of education (OR 1.48 [95% CI 1.37 to 1.61]) and the elderly (OR 7.49 [95% CI 6.53 to 8.59]) had a greater risk for the disease. CONCLUSIONS CHIK stands as an emerging public health problem in Brazil since its introduction in 2014. Espírito Santo suffered a substantial epidemic in 2020, possibly due to outbreaks in neighbouring states. The population at risk should be prioritized in healthcare, considering the morbidity potential of the disease.
Collapse
Affiliation(s)
- Marcus Vinicius Salvador Clipes
- Post-Graduate Program of Infectious Diseases, Federal University of Espirito Santo, Vitória, Marechal Campos Avenue, 1468, 29047-105, Espírito Santo, Brazil
| | - Creuza Rachel Vicente
- Post-Graduate Program of Infectious Diseases, Federal University of Espirito Santo, Vitória, Marechal Campos Avenue, 1468, 29047-105, Espírito Santo, Brazil
| | - Theresa Cristina Cardoso da Silva
- Special Nucleus of Epidemiologic Surveillance from the State Department of Health, Vitória, Marechal Mascarenha de Moraes Avenue, 29050-755, Espírito Santo, Brazil
| | - Lilyan Correia Resende
- Post-Graduate Program of Infectious Diseases, Federal University of Espirito Santo, Vitória, Marechal Campos Avenue, 1468, 29047-105, Espírito Santo, Brazil
| | - Crispim Cerutti Junior
- Post-Graduate Program of Infectious Diseases, Federal University of Espirito Santo, Vitória, Marechal Campos Avenue, 1468, 29047-105, Espírito Santo, Brazil
| |
Collapse
|
26
|
da Silva MOL, Figueiredo CM, Neris RLS, Guimarães-Andrade IP, Gavino-Leopoldino D, Miler-da-Silva LL, Valença HDM, Ladislau L, de Lima CVF, Coccarelli FM, Benjamim CF, Assunção-Miranda I. Chikungunya and Mayaro Viruses Induce Chronic Skeletal Muscle Atrophy Triggered by Pro-Inflammatory and Oxidative Response. Int J Mol Sci 2024; 25:8909. [PMID: 39201595 PMCID: PMC11354814 DOI: 10.3390/ijms25168909] [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: 02/23/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 09/02/2024] Open
Abstract
Chikungunya (CHIKV) and Mayaro (MAYV) viruses are arthritogenic alphaviruses that promote an incapacitating and long-lasting inflammatory muscle-articular disease. Despite studies pointing out the importance of skeletal muscle (SkM) in viral pathogenesis, the long-term consequences on its physiology and the mechanism of persistence of symptoms are still poorly understood. Combining molecular, morphological, nuclear magnetic resonance imaging, and histological analysis, we conduct a temporal investigation of CHIKV and MAYV replication in a wild-type mice model, focusing on the impact on SkM composition, structure, and repair in the acute and late phases of infection. We found that viral replication and induced inflammation promote a rapid loss of muscle mass and reduction in fiber cross-sectional area by upregulation of muscle-specific E3 ubiquitin ligases MuRF1 and Atrogin-1 expression, both key regulators of SkM fibers atrophy. Despite a reduction in inflammation and clearance of infectious viral particles, SkM atrophy persists until 30 days post-infection. The genomic CHIKV and MAYV RNAs were still detected in SkM in the late phase, along with the upregulation of chemokines and anti-inflammatory cytokine expression. In agreement with the involvement of inflammatory mediators on induced atrophy, the neutralization of TNF and a reduction in oxidative stress using monomethyl fumarate, an agonist of Nrf2, decreases atrogen expression and atrophic fibers while increasing weight gain in treated mice. These data indicate that arthritogenic alphavirus infection could chronically impact body SkM composition and also harm repair machinery, contributing to a better understanding of mechanisms of arthritogenic alphavirus pathogenesis and with a description of potentially new targets of therapeutic intervention.
Collapse
Affiliation(s)
- Mariana Oliveira Lopes da Silva
- Department of Virology, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (M.O.L.d.S.); (C.M.F.); (R.L.S.N.); (I.P.G.-A.); (D.G.-L.)
| | - Camila Menezes Figueiredo
- Department of Virology, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (M.O.L.d.S.); (C.M.F.); (R.L.S.N.); (I.P.G.-A.); (D.G.-L.)
| | - Rômulo Leão Silva Neris
- Department of Virology, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (M.O.L.d.S.); (C.M.F.); (R.L.S.N.); (I.P.G.-A.); (D.G.-L.)
| | - Iris Paula Guimarães-Andrade
- Department of Virology, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (M.O.L.d.S.); (C.M.F.); (R.L.S.N.); (I.P.G.-A.); (D.G.-L.)
| | - Daniel Gavino-Leopoldino
- Department of Virology, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (M.O.L.d.S.); (C.M.F.); (R.L.S.N.); (I.P.G.-A.); (D.G.-L.)
| | - Leonardo Linhares Miler-da-Silva
- Department of Virology, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (M.O.L.d.S.); (C.M.F.); (R.L.S.N.); (I.P.G.-A.); (D.G.-L.)
| | - Helber da Maia Valença
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (H.d.M.V.)
| | - Leandro Ladislau
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (H.d.M.V.)
| | - Caroline Victorino Felix de Lima
- National Center for Structural Biology and Bioimaging (CENABio), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (C.V.F.d.L.); (F.M.C.)
- Instituto D’Or de Pesquisa e Ensino, Rio de Janeiro 22281-100, Brazil
| | - Fernanda Meireles Coccarelli
- National Center for Structural Biology and Bioimaging (CENABio), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (C.V.F.d.L.); (F.M.C.)
- Instituto D’Or de Pesquisa e Ensino, Rio de Janeiro 22281-100, Brazil
| | - Claudia Farias Benjamim
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil;
| | - Iranaia Assunção-Miranda
- Department of Virology, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (M.O.L.d.S.); (C.M.F.); (R.L.S.N.); (I.P.G.-A.); (D.G.-L.)
| |
Collapse
|
27
|
de Oliveira Souza R, Duarte Júnior JWB, Della Casa VS, Santoro Rosa D, Renia L, Claser C. Unraveling the complex interplay: immunopathology and immune evasion strategies of alphaviruses with emphasis on neurological implications. Front Cell Infect Microbiol 2024; 14:1421571. [PMID: 39211797 PMCID: PMC11358129 DOI: 10.3389/fcimb.2024.1421571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/09/2024] [Indexed: 09/04/2024] Open
Abstract
Arthritogenic alphaviruses pose a significant public health concern due to their ability to cause joint inflammation, with emerging evidence of potential neurological consequences. In this review, we examine the immunopathology and immune evasion strategies employed by these viruses, highlighting their complex mechanisms of pathogenesis and neurological implications. We delve into how these viruses manipulate host immune responses, modulate inflammatory pathways, and potentially establish persistent infections. Further, we explore their ability to breach the blood-brain barrier, triggering neurological complications, and how co-infections exacerbate neurological outcomes. This review synthesizes current research to provide a comprehensive overview of the immunopathological mechanisms driving arthritogenic alphavirus infections and their impact on neurological health. By highlighting knowledge gaps, it underscores the need for research to unravel the complexities of virus-host interactions. This deeper understanding is crucial for developing targeted therapies to address both joint and neurological manifestations of these infections.
Collapse
Affiliation(s)
- Raquel de Oliveira Souza
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | | | - Victória Simões Della Casa
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Daniela Santoro Rosa
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Laurent Renia
- ASTAR Infectious Diseases Labs (ASTAR ID Labs), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Carla Claser
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| |
Collapse
|
28
|
Hick TAH, Geertsema C, Nguyen W, Bishop CR, van Oosten L, Abbo SR, Dumenil T, van Kuppeveld FJM, Langereis MA, Rawle DJ, Tang B, Yan K, van Oers MM, Suhrbier A, Pijlman GP. Safety concern of recombination between self-amplifying mRNA vaccines and viruses is mitigated in vivo. Mol Ther 2024; 32:2519-2534. [PMID: 38894543 PMCID: PMC11405153 DOI: 10.1016/j.ymthe.2024.06.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 04/02/2024] [Accepted: 06/14/2024] [Indexed: 06/21/2024] Open
Abstract
Self-amplifying mRNA (SAM) vaccines can be rapidly deployed in the event of disease outbreaks. A legitimate safety concern is the potential for recombination between alphavirus-based SAM vaccines and circulating viruses. This theoretical risk needs to be assessed in the regulatory process for SAM vaccine approval. Herein, we undertake extensive in vitro and in vivo assessments to explore recombination between SAM vaccine and a wide selection of alphaviruses and a coronavirus. SAM vaccines were found to effectively limit alphavirus co-infection through superinfection exclusion, although some co-replication was still possible. Using sensitive cell-based assays, replication-competent alphavirus chimeras were generated in vitro as a result of rare, but reproducible, RNA recombination events. The chimeras displayed no increased fitness in cell culture. Viable alphavirus chimeras were not detected in vivo in C57BL/6J, Rag1-/- and Ifnar-/- mice, in which high levels of SAM vaccine and alphavirus co-replicated in the same tissue. Furthermore, recombination between a SAM-spike vaccine and a swine coronavirus was not observed. In conclusion we state that although the ability of SAM vaccines to recombine with alphaviruses might be viewed as an environmental safety concern, several key factors substantially mitigate against in vivo emergence of chimeric viruses from SAM vaccine recipients.
Collapse
Affiliation(s)
- Tessy A H Hick
- Laboratory of Virology, Wageningen University and Research, Wageningen, the Netherlands
| | - Corinne Geertsema
- Laboratory of Virology, Wageningen University and Research, Wageningen, the Netherlands
| | - Wilson Nguyen
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Cameron R Bishop
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Linda van Oosten
- Laboratory of Virology, Wageningen University and Research, Wageningen, the Netherlands
| | - Sandra R Abbo
- Laboratory of Virology, Wageningen University and Research, Wageningen, the Netherlands
| | - Troy Dumenil
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Frank J M van Kuppeveld
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, the Netherlands
| | - Martijn A Langereis
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, the Netherlands
| | - Daniel J Rawle
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Bing Tang
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Kexin Yan
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Monique M van Oers
- Laboratory of Virology, Wageningen University and Research, Wageningen, the Netherlands
| | - Andreas Suhrbier
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia; Global Virus Network Centre of Excellence, Australian Infectious Diseases Research Centre, Brisbane, QLD 4072 and 4029, Australia.
| | - Gorben P Pijlman
- Laboratory of Virology, Wageningen University and Research, Wageningen, the Netherlands.
| |
Collapse
|
29
|
Dunagan MM, Dábilla N, McNinch C, Brenchley JM, Dolan PT, Fox JM. Activating FcγRs on monocytes are necessary for optimal Mayaro virus clearance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.23.604823. [PMID: 39149309 PMCID: PMC11326306 DOI: 10.1101/2024.07.23.604823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Mayaro virus (MAYV) is an emerging arbovirus. Previous studies have shown antibody Fc effector functions are critical for optimal monoclonal antibody-mediated protection against alphaviruses; however, the requirement of Fc gamma receptors (FcγRs) for protection during natural infection has not been evaluated. Here, we showed mice lacking activating FcγRs (FcRγ-/-) developed prolonged clinical disease with more virus in joint-associated tissues. Viral clearance was associated with anti-MAYV cell surface binding rather than neutralizing antibodies. Lack of Fc-FcγR engagement increased the number of monocytes through chronic timepoints. Single cell RNA sequencing showed elevated levels of pro-inflammatory monocytes in joint-associated tissue with increased MAYV RNA present in FcRγ-/- monocytes and macrophages. Transfer of FcRγ-/- monocytes into wild type animals was sufficient to increase virus in joint-associated tissue. Overall, this study suggests that engagement of antibody Fc with activating FcγRs promotes protective responses during MAYV infection and prevents monocytes from being potential targets of infection.
Collapse
Affiliation(s)
- Megan M. Dunagan
- Emerging Virus Immunity Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Nathânia Dábilla
- Quantitative Virology and Evolution Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Colton McNinch
- Bioinformatics and Computational Bioscience Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Jason M. Brenchley
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Patrick T. Dolan
- Quantitative Virology and Evolution Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Julie M. Fox
- Emerging Virus Immunity Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
30
|
Freppel W, Lim EX, Rudd PA, Herrero LJ. Synoviocytes assist in modulating the effect of Ross River virus infection in micromass-cultured primary human chondrocytes. J Med Microbiol 2024; 73:001859. [PMID: 39028255 PMCID: PMC11316548 DOI: 10.1099/jmm.0.001859] [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: 03/02/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
Introduction. Ross River virus (RRV) is a mosquito-borne virus prevalent in Australia and the islands of the South Pacific, where it causes an arthritogenic illness with a hallmark feature of severe joint pain. The joint space is a unique microenvironment that contains cartilage and synovial fluid. Chondrocytes and synoviocytes are crucial components of the joint space and are known targets of RRV infection.Hypothesis/Gap statement. Understanding the relationship between synoviocytes and chondrocytes during RRV infection will provide further insights into RRV-induced joint pathology.Methodology. To better understand the unique dynamics of these cells during RRV infection, we used primary chondrocytes cultured in physiologically relevant micromasses. We then directly infected micromass chondrocytes or infected primary fibroblast-like synoviocytes (FLS), co-cultured with micromass chondrocytes. Micromass cultures and supernatants were collected and analysed for viral load with a PCR array of target genes known to play a role in arthritis.Results. We show that RRV through direct or secondary infection in micromass chondrocytes modulates the expression of cellular factors that likely contribute to joint inflammation and disease pathology, as well as symptoms such as pain. More importantly, while we show that RRV can infect micromass-cultured chondrocytes via FLS infection, FLS themselves affect the regulation of cellular genes known to contribute to arthritis.Conclusion. Single-cell culture systems lack the complexity of in vivo systems, and understanding the interaction between cell populations is crucial for deciphering disease pathology, including for the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Wesley Freppel
- Institute for Biomedicine and Glycomics, Gold Coast Campus, Griffith University, Southport, QLD 4222, Australia
| | - Elisa X.Y. Lim
- Institute for Biomedicine and Glycomics, Gold Coast Campus, Griffith University, Southport, QLD 4222, Australia
| | - Penny A. Rudd
- Institute for Biomedicine and Glycomics, Gold Coast Campus, Griffith University, Southport, QLD 4222, Australia
| | - Lara J. Herrero
- Institute for Biomedicine and Glycomics, Gold Coast Campus, Griffith University, Southport, QLD 4222, Australia
| |
Collapse
|
31
|
Ghoshal A, Magalhães ÁF, Asressu KH, Hossain MA, Todd MH, Willson TM. Identification of Dihydropyrazolo[1,5- a]pyrazin-4(5 H)-ones as Cyclic Products of β-Amidomethyl Vinyl Sulfone Alphavirus Cysteine Protease Inhibitors. Pharmaceuticals (Basel) 2024; 17:836. [PMID: 39065687 PMCID: PMC11279629 DOI: 10.3390/ph17070836] [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: 06/10/2024] [Revised: 06/18/2024] [Accepted: 06/23/2024] [Indexed: 07/28/2024] Open
Abstract
Optimized syntheses of (E)-5-(2-ethoxyphenyl)-N-(3-(methylsulfonyl)allyl)-1H-pyrazole-3-carboxamide (RA-0002034, 1), a promising antiviral covalent cysteine protease inhibitor lead, were developed. The syntheses avoid the contamination of 1 with the inactive cyclic dihydropyrazolo[1,5-a]pyrazin-4(5H)-one 2, which is formed by the intramolecular aza-Michael reaction of the vinyl sulfone warhead under basic conditions and slowly at pH 7.4 in phosphate buffer. The pure cysteine protease inhibitor 1 could be synthesized using either modified amide coupling conditions or through the introduction of a MOM-protecting group and was stable as a TFA or HCl salt. Although acyclic 1 demonstrated poor pharmacokinetics with high in vivo clearance in mice, inactive cyclic 2 showed improved plasma exposure. The potential use of cyclic dihydropyrazolo[1,5-a]pyrazin-4(5H)-ones as prodrugs for the acyclic β-amidomethyl vinyl sulfone warhead was demonstrated by GSH capture experiments with an analog of 2.
Collapse
Affiliation(s)
- Anirban Ghoshal
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Álvaro F. Magalhães
- Structural Genomics Consortium, Department of Pharmaceutical and Biological Chemistry, School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Kesatebrhan Haile Asressu
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mohammad Anwar Hossain
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matthew H. Todd
- Structural Genomics Consortium, Department of Pharmaceutical and Biological Chemistry, School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Timothy M. Willson
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
32
|
Ghoshal A, Asressu KH, Hossain MA, Brown PJ, Merten EM, Sears JD, Perveen S, Pearce KH, Popov KI, Moorman NJ, Heise MT, Willson TM. Structure Activity of β-Amidomethyl Vinyl Sulfones as Covalent Inhibitors of Chikungunya nsP2 Cysteine Protease with Anti-alphavirus Activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598722. [PMID: 38915519 PMCID: PMC11195264 DOI: 10.1101/2024.06.12.598722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Despite their widespread impact on human health there are no approved drugs for combating alphavirus infections. The heterocyclic β-aminomethyl vinyl sulfone RA-0002034 (1a) is a potent irreversible covalent inhibitor of the alphavirus nsP2 cysteine protease with broad spectrum antiviral activity. Analogs of 1a that varied each of three regions of the molecule were synthesized to establish structure-activity relationships for inhibition of Chikungunya (CHIKV) nsP2 protease and viral replication. The covalent warhead was highly sensitive to modifications of the sulfone or vinyl substituents. However, numerous alterations to the core 5-membered heterocycle and its aryl substituent were well tolerated and several analogs were identified that enhanced CHIKV nsP2 binding. For example, the 4-cyanopyrazole analog 8d exhibited a kinact /Ki ratio >10,000 M-1s-1. 3-Arylisoxazole was identified an isosteric replacement for the 5-membered heterocycle, which circumvented the intramolecular cyclization that complicated the synthesis of pyrazole-based inhibitors like 1a. The accumulated structure-activity data was used to build a ligand-based model of the enzyme active site, which can be used to guide the design of covalent nsP2 protease inhibitors as potential therapeutics against alphaviruses.
Collapse
Affiliation(s)
- Anirban Ghoshal
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kesatebrhan Haile Asressu
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mohammad Anwar Hossain
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Peter J. Brown
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Eric M. Merten
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - John D. Sears
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Sumera Perveen
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, M5G 1L7, Canada
| | - Kenneth H. Pearce
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Konstantin I. Popov
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nathaniel J. Moorman
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Mark T. Heise
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Timothy M. Willson
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- READDI AViDD Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
33
|
Baker H, Amaral JK, Schoen RT. Management of postinfectious inflammatory arthritis. Curr Opin Rheumatol 2024; 36:155-162. [PMID: 38411201 DOI: 10.1097/bor.0000000000001009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
PURPOSE OF REVIEW Postinfectious inflammatory arthritis can result from various pathogens, including bacteria, viruses, fungi, and parasites. Prompt identification and treatment of acute infection is vital, but some cases progress to chronic arthritis despite successful treatment of infection. Postinfectious inflammatory arthritis varies from mild, self-limited arthralgia to severe, refractory arthritis, necessitating ongoing disease-modifying treatment. This review explores the spectrum of postinfectious inflammatory arthritis to provide insights into effective management. RECENT FINDINGS Research continues regarding the benefit of antimicrobial therapy, beyond treatment of the acute infection, to diminish the severity of postinfectious inflammatory arthritis. Following treatment of acute infection, most cases are self-limited so treatment is symptomatic. However, a difficult-to-predict fraction of cases develop chronic postinfectious inflammatory arthritis that can be challenging to manage. Recently, as more biologic, and targeted synthetic DMARDs have become available, treatment options have expanded. SUMMARY In this article, we use the term 'postinfectious inflammatory arthritis' rather than 'reactive arthritis' because it describes a broader spectrum of diseases and emphasizes the common pathogenesis of a postinfectious inflammatory process. We summarize the conventional therapies and recent management developments for the most frequently encountered postinfectious inflammatory arthritides.
Collapse
Affiliation(s)
- Hailey Baker
- Section of Rheumatology, Allergy, and Immunology, Yale School of Medicine
| | - J Kennedy Amaral
- Institute of Diagnostic Medicine of Cariri, Juazeiro do Norte, Ceará, Brazil
| | - Robert T Schoen
- Section of Rheumatology, Allergy, and Immunology, Yale School of Medicine
| |
Collapse
|
34
|
Rawat SK, Kale D, Nema S, Nema RK, Gupta S, Khadanga S, Biswas D. Detectable Viremia at Presentation Is a Predictor of Disease Severity in Chikungunya. Cureus 2024; 16:e58188. [PMID: 38741833 PMCID: PMC11089836 DOI: 10.7759/cureus.58188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2024] [Indexed: 05/16/2024] Open
Abstract
Background Chikungunya is a mosquito-borne re-emerging disease that has caused a significant number of outbreaks recently in diverse geographic settings across the globe. It leads to severe debilitating illness in a significant proportion of persons who are infected. Measures to limit the impact produced by recurrent outbreaks of the disease are limited and there is an urgent clinical need for early identification of those predisposed to develop severe disease. A comprehensive understanding regarding the proportion of individuals predisposed to developing severe disease is lacking as its correlation with detectable viremia is hinted at by some studies. In this context, we hypothesized that detectable viremia reflected in the diagnostic RT-PCR assay could be significantly associated with the development of severe disease in Chikungunya among those diagnosed on the basis of seroconversion. Our study aims to confirm the same in relation to disease severity among the suspected patients of Chikungunya in the setting of a tertiary care center. Methods In a prospective observational study at a tertiary care center, a total number of 1021 Chikungunya suspects presenting within seven days of illness were screened with Chikungunya Virus IgM ELISA from 2021 to 2023. Those having positive IgM results were further tested with RT-PCR in a blinded manner. According to the information entered into the predesigned form and the hospital follow-up/discharge data, the cases where symptoms like fever and joint pain persisted beyond two weeks were classified as severe versus those resolving within two weeks as mild. The patients in each group were compared for their clinical symptoms and association with the disease severity with detectable viremia (RT-PCR positivity). Results We identified a total of 178 (17.4%) lab-confirmed Chikungunya IgM-positive cases amongst the recruited patients. Here a total of 31 (18.9%) cases could be classified as severe and 133 (74.7%) as mild illness, the remaining 14 patients were excluded from analysis due to insufficient clinical data. Severe illness was significantly higher in elderly individuals belonging to more than 60 years (p = 0.01). Viremia was detected in 16 (9%), those with detectable viremia had higher odds (OR = 4.1) of manifesting as severe disease. Among the severe cases, the proportion of cases with RT-PCR positivity (8, 25.8%) at presentation was significantly higher (P = 0.01) versus those who presented with mild disease (7, 5.5%). Conclusion Our study reveals a correlation between detectable viremia in Chikungunya virus (CHIKV) patients and an increased risk of manifesting into a severe disease, where severe cases exhibited a significantly higher proportion of viremia, indicated by RT-PCR positivity. This study hints at the presence of viremia, joint symptoms, and elderly age as potentially useful clinical predictors of disease outcomes, these may serve as indicators for closer monitoring among individuals seeking medical attention due to Chikungunya infection. However, we need to validate these findings in future longitudinal studies incorporating multiple, time-bound follow-up data on clinical outcomes, viral titers, and its long-term complications.
Collapse
Affiliation(s)
- Sumit K Rawat
- Microbiology, Bundelkhand Medical College, Sagar, IND
- Microbiology, All India Institute of Medical Sciences, Bhopal, Bhopal, IND
| | - Dipesh Kale
- Microbiology, All India Institute of Medical Sciences, Bhopal, Bhopal, IND
| | - Shashwati Nema
- Microbiology, All India Institute of Medical Sciences, Bhopal, Bhopal, IND
| | - Ram K Nema
- Molecular Biology, Indian Council of Medical Research (ICMR) National Institute for Research in Environmental Health, Bhopal, IND
| | - Sudheer Gupta
- Next-Generation Sequencing (NGS) & Bioinformatics Division, 3B Blackbio Dx Ltd., Bhopal, IND
| | - Sagar Khadanga
- Internal Medicine, All India Institute of Medical Sciences, Bhopal, Bhopal, IND
| | - Debasis Biswas
- Microbiology, All India Institute of Medical Sciences, Bhopal, Bhopal, IND
| |
Collapse
|
35
|
Wang Y, Xu Z, Zhang H, Zhou Y, Cao J, Zhang Y, Wang Z, Zhou J. Towards modelling tick-virus interactions using the weakly pathogenic Sindbis virus: Evidence that ticks are competent vectors. Front Cell Infect Microbiol 2024; 14:1334351. [PMID: 38567020 PMCID: PMC10985168 DOI: 10.3389/fcimb.2024.1334351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/27/2024] [Indexed: 04/04/2024] Open
Abstract
Most tick-borne viruses (TBVs) are highly pathogenic and require high biosecurity, which severely limits their study. We found that Sindbis virus (SINV), predominantly transmitted by mosquitoes, can replicate in ticks and be subsequently transmitted, with the potential to serve as a model for studying tick-virus interactions. We found that both larval and nymphal stages of Rhipicephalus haemaphysaloides can be infected with SINV-wild-type (WT) when feeding on infected mice. SINV replicated in two species of ticks (R. haemaphysaloides and Hyalomma asiaticum) after infecting them by microinjection. Injection of ticks with SINV expressing enhanced Green Fluorescent Protein (eGFP) revealed that SINV-eGFP specifically aggregated in the tick midguts for replication. During blood-feeding, SINV-eGFP migrated from the midguts to the salivary glands and was transmitted to a new host. SINV infection caused changes in expression levels of tick genes related to immune responses, substance transport and metabolism, cell growth and death. SINV mainly induced autophagy during the early stage of infection; with increasing time of infection, the level of autophagy decreased, while the level of apoptosis increased. During the early stages of infection, the transcript levels of immune-related genes were significantly upregulated, and then decreased. In addition, SINV induced changes in the transcription levels of some functional genes that play important roles in the interactions between ticks and tick-borne pathogens. These results confirm that the SINV-based transmission model between ticks, viruses, and mammals can be widely used to unravel the interactions between ticks and viruses.
Collapse
Affiliation(s)
- Yanan Wang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Zhengmao Xu
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Houshuang Zhang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yongzhi Zhou
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Jie Cao
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yuqiang Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Zedong Wang
- Center of Infectious Diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Jilin, China
| | - Jinlin Zhou
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| |
Collapse
|
36
|
Mehta D, Chaudhary S, Sunil S. Oxidative stress governs mosquito innate immune signalling to reduce chikungunya virus infection in Aedes-derived cells. J Gen Virol 2024; 105. [PMID: 38488850 DOI: 10.1099/jgv.0.001966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024] Open
Abstract
Arboviruses such as chikungunya, dengue and zika viruses cause debilitating diseases in humans. The principal vector species that transmits these viruses is the Aedes mosquito. Lack of substantial knowledge of the vector species hinders the advancement of strategies for controlling the spread of arboviruses. To supplement our information on mosquitoes' responses to virus infection, we utilized Aedes aegypti-derived Aag2 cells to study changes at the transcriptional level during infection with chikungunya virus (CHIKV). We observed that genes belonging to the redox pathway were significantly differentially regulated. Upon quantifying reactive oxygen species (ROS) in the cells during viral infection, we further discovered that ROS levels are considerably higher during the early hours of infection; however, as the infection progresses, an increase in antioxidant gene expression suppresses the oxidative stress in cells. Our study also suggests that ROS is a critical regulator of viral replication in cells and inhibits intracellular and extracellular viral replication by promoting the Rel2-mediated Imd immune signalling pathway. In conclusion, our study provides evidence for a regulatory role of oxidative stress in infected Aedes-derived cells.
Collapse
Affiliation(s)
- Divya Mehta
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Sakshi Chaudhary
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Sujatha Sunil
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| |
Collapse
|
37
|
McMahon R, Fuchs U, Schneider M, Hadl S, Hochreiter R, Bitzer A, Kosulin K, Koren M, Mader R, Zoihsl O, Wressnigg N, Dubischar K, Buerger V, Eder-Lingelbach S, Jaramillo JC. A randomized, double-blinded Phase 3 study to demonstrate lot-to-lot consistency and to confirm immunogenicity and safety of the live-attenuated chikungunya virus vaccine candidate VLA1553 in healthy adults†. J Travel Med 2024; 31:taad156. [PMID: 38091981 PMCID: PMC10911060 DOI: 10.1093/jtm/taad156] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 03/05/2024]
Abstract
BACKGROUND The global spread of the chikungunya virus (CHIKV) increases the exposure risk for individuals travelling to or living in endemic areas. This Phase 3 study was designed to demonstrate manufacturing consistency between three lots of the single shot live-attenuated CHIKV vaccine VLA1553, and to confirm the promising immunogenicity and safety data obtained in previous trials. METHODS This randomized, double-blinded, lot-to-lot consistency, Phase 3 study, assessed immunogenicity and safety of VLA1553 in 408 healthy adults (18-45 years) in 12 sites across the USA. The primary endpoint was a comparison of the geometric mean titre (GMT) ratios of CHIKV-specific neutralizing antibodies between three VLA1553 lots at 28 days post-vaccination. Secondary endpoints included immunogenicity and safety over 6 months post-vaccination. RESULTS GMTs were comparable between the lots meeting the acceptance criteria for equivalence. The average GMT (measured by 50% CHIKV micro plaque neutralization test; μPRNT50) peaked with 2643 at 28 days post-vaccination and decreased to 709 at 6 months post-vaccination. An excellent seroresponse rate (defined as μPRNT50 titre ≥ 150 considered protective) was achieved in 97.8% of participants at 28 days post-vaccination and still persisted in 96% at 6 months after vaccination. Upon VLA1553 immunization, 72.5% of participants experienced adverse events (AEs), without significant differences between lots (related solicited systemic AE: 53.9% of participants; related solicited local AE: 19.4%). Overall, AEs were mostly mild or moderate and resolved without sequela, usually within 3 days. With 3.9% of participants experiencing severe AEs, 2.7% were classified as related, whereas none of the six reported serious adverse events was related to the administration of VLA1553. CONCLUSIONS All three lots of VLA1553 recapitulated the safety and immunogenicity profiles of a preceding Phase 3 study, fulfilling pre-defined consistency requirements. These results highlight the manufacturability of VLA1553, a promising vaccine for the prevention of CHIKV disease for those living in or travelling to endemic areas.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Michael Koren
- Walter Reed Army Institute of Research, Bethesda, MD, USA
| | - Robert Mader
- CRETA GmbH, Campus Vienna Biocenter 3, 1030 Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
38
|
Chykunova Y, Plewka J, Wilk P, Torzyk K, Sienczyk M, Dubin G, Pyrc K. Autoinhibition of suicidal capsid protease from O'nyong'nyong virus. Int J Biol Macromol 2024; 262:130136. [PMID: 38354926 DOI: 10.1016/j.ijbiomac.2024.130136] [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: 10/10/2023] [Revised: 01/10/2024] [Accepted: 02/11/2024] [Indexed: 02/16/2024]
Abstract
Alphaviruses pose a significant threat to public health. Capsid protein encoded in the alphaviral genomes constitutes an interesting therapy target, as it also serves as a protease (CP). Remarkably, it undergoes autoproteolysis, leading to the generation of the C-terminal tryptophan that localizes to the active pocket, deactivating the enzyme. Lack of activity hampers the viral replication cycle, as the virus is not capable of producing the infectious progeny. We investigated the structure and function of the CP encoded in the genome of O'nyong'nyong virus (ONNV), which has instigated outbreaks in Africa. Our research provides a high-resolution crystal structure of the ONNV CP in its active state and evaluates the enzyme's activity. Furthermore, we demonstrated a dose-dependent reduction in ONNV CP proteolytic activity when exposed to indole, suggesting that tryptophan analogs may be a promising basis for developing small molecule inhibitors. It's noteworthy that the capsid protease plays an essential role in virus assembly, binding viral glycoproteins through its glycoprotein-binding hydrophobic pocket. We showed that non-aromatic cyclic compounds like dioxane disrupt this vital interaction. Our findings provide deeper insights into ONNV's biology, and we believe they will prove instrumental in guiding the development of antiviral strategies against arthritogenic alphaviruses.
Collapse
Affiliation(s)
- Yuliya Chykunova
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland; Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Jacek Plewka
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Piotr Wilk
- Structural Biology Core Facility, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland
| | - Karolina Torzyk
- Wroclaw University of Science and Technology, Faculty of Chemistry, Division of Medicinal Chemistry and Microbiology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Marcin Sienczyk
- Wroclaw University of Science and Technology, Faculty of Chemistry, Division of Medicinal Chemistry and Microbiology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Grzegorz Dubin
- Protein Crystallography Research Group, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland.
| | - Krzysztof Pyrc
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland.
| |
Collapse
|
39
|
Zhai X, Li X, Veit M, Wang N, Wang Y, Merits A, Jiang Z, Qin Y, Zhang X, Qi K, Jiao H, He WT, Chen Y, Mao Y, Su S. LDLR is used as a cell entry receptor by multiple alphaviruses. Nat Commun 2024; 15:622. [PMID: 38245515 PMCID: PMC10799924 DOI: 10.1038/s41467-024-44872-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 01/04/2024] [Indexed: 01/22/2024] Open
Abstract
Alphaviruses are arboviruses transmitted by mosquitoes and are pathogenic to humans and livestock, causing a substantial public health burden. So far, several receptors have been identified for alphavirus entry; however, they cannot explain the broad host range and tissue tropism of certain alphaviruses, such as Getah virus (GETV), indicating the existence of additional receptors. Here we identify the evolutionarily conserved low-density lipoprotein receptor (LDLR) as a new cell entry factor for GETV, Semliki Forest virus (SFV), Ross River virus (RRV) and Bebaru virus (BEBV). Ectopic expression of LDLR facilitates cellular binding and internalization of GETV, which is mediated by the interaction between the E2-E1 spike of GETV and the ligand-binding domain (LBD) of LDLR. Antibodies against LBD block GETV infection in cultured cells. In addition, the GST-LBD fusion protein inhibits GETV infection both in vitro and in vivo. Notably, we identify the key amino acids in LDLR-LBD that played a crucial role in viral entry; specific mutations in the CR4 and CR5 domain of LDLR-LBD reduce viral entry to cells by more than 20-fold. These findings suggest that targeting the LDLR-LBD could be a potential strategy for the development of antivirals against multiple alphaviruses.
Collapse
Affiliation(s)
- Xiaofeng Zhai
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiaoling Li
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Michael Veit
- Institute for Virology, Center for Infection Medicine, Veterinary Faculty, Free University Berlin, Berlin, Germany
| | - Ningning Wang
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yu Wang
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Andres Merits
- Institute of Bioengineering, University of Tartu, Nooruse Street 1, 50411, Tartu, Estonia
| | - Zhiwen Jiang
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yan Qin
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiaoguang Zhang
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Kaili Qi
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Houqi Jiao
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Wan-Ting He
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ye Chen
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Yang Mao
- School of Pharmaceutical Sciences and National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou, China.
| | - Shuo Su
- Academy for Advanced Interdisciplinary Studies, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.
| |
Collapse
|
40
|
Adams LJ, Raju S, Ma H, Gilliland T, Reed DS, Klimstra WB, Fremont DH, Diamond MS. Structural and functional basis of VLDLR usage by Eastern equine encephalitis virus. Cell 2024; 187:360-374.e19. [PMID: 38176410 PMCID: PMC10843625 DOI: 10.1016/j.cell.2023.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 10/06/2023] [Accepted: 11/28/2023] [Indexed: 01/06/2024]
Abstract
The very-low-density lipoprotein receptor (VLDLR) comprises eight LDLR type A (LA) domains and supports entry of distantly related alphaviruses, including Eastern equine encephalitis virus (EEEV) and Semliki Forest virus (SFV). Here, by resolving multiple cryo-electron microscopy structures of EEEV-VLDLR complexes and performing mutagenesis and functional studies, we show that EEEV uses multiple sites (E1/E2 cleft and E2 A domain) to engage more than one LA domain simultaneously. However, no single LA domain is necessary or sufficient to support efficient EEEV infection. Whereas all EEEV strains show conservation of two VLDLR-binding sites, the EEEV PE-6 strain and a few other EEE complex members feature a single amino acid substitution that enables binding of LA domains to an additional site on the E2 B domain. These structural and functional analyses informed the design of a minimal VLDLR decoy receptor that neutralizes EEEV infection and protects mice from lethal challenge.
Collapse
Affiliation(s)
- Lucas J Adams
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Saravanan Raju
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hongming Ma
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Theron Gilliland
- The Center for Vaccine Research and Department of Immunology, The University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Douglas S Reed
- The Center for Vaccine Research and Department of Immunology, The University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - William B Klimstra
- The Center for Vaccine Research and Department of Immunology, The University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Daved H Fremont
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Michael S Diamond
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
41
|
Ma H, Adams LJ, Raju S, Sariol A, Kafai NM, Janova H, Klimstra WB, Fremont DH, Diamond MS. The low-density lipoprotein receptor promotes infection of multiple encephalitic alphaviruses. Nat Commun 2024; 15:246. [PMID: 38172096 PMCID: PMC10764363 DOI: 10.1038/s41467-023-44624-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
Members of the low-density lipoprotein receptor (LDLR) family, including LDLRAD3, VLDLR, and ApoER2, were recently described as entry factors for different alphaviruses. However, based on studies with gene edited cells and knockout mice, blockade or abrogation of these receptors does not fully inhibit alphavirus infection, indicating the existence of additional uncharacterized entry factors. Here, we perform a CRISPR-Cas9 genome-wide loss-of-function screen in mouse neuronal cells with a chimeric alphavirus expressing the Eastern equine encephalitis virus (EEEV) structural proteins and identify LDLR as a candidate receptor. Expression of LDLR on the surface of neuronal or non-neuronal cells facilitates binding and infection of EEEV, Western equine encephalitis virus, and Semliki Forest virus. Domain mapping and binding studies reveal a low-affinity interaction with LA domain 3 (LA3) that can be enhanced by concatenation of LA3 repeats. Soluble decoy proteins with multiple LA3 repeats inhibit EEEV infection in cell culture and in mice. Our results establish LDLR as a low-affinity receptor for multiple alphaviruses and highlight a possible path for developing inhibitors that could mitigate infection and disease.
Collapse
Affiliation(s)
- Hongming Ma
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Lucas J Adams
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Saravanan Raju
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Alan Sariol
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Natasha M Kafai
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hana Janova
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - William B Klimstra
- The Center for Vaccine Research and Department of Immunology, The University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Daved H Fremont
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
| |
Collapse
|
42
|
Celone M, Beeman S, Han BA, Potter AM, Pecor DB, Okech B, Pollett S. Understanding transmission risk and predicting environmental suitability for Mayaro Virus in Central and South America. PLoS Negl Trop Dis 2024; 18:e0011859. [PMID: 38194417 PMCID: PMC10775973 DOI: 10.1371/journal.pntd.0011859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 12/12/2023] [Indexed: 01/11/2024] Open
Abstract
Mayaro virus (MAYV) is a mosquito-borne Alphavirus that is widespread in South America. MAYV infection often presents with non-specific febrile symptoms but may progress to debilitating chronic arthritis or arthralgia. Despite the pandemic threat of MAYV, its true distribution remains unknown. The objective of this study was to clarify the geographic distribution of MAYV using an established risk mapping framework. This consisted of generating evidence consensus scores for MAYV presence, modeling the potential distribution of MAYV in select countries across Central and South America, and estimating the population residing in areas suitable for MAYV transmission. We compiled a georeferenced compendium of MAYV occurrence in humans, animals, and arthropods. Based on an established evidence consensus framework, we integrated multiple information sources to assess the total evidence supporting ongoing transmission of MAYV within each country in our study region. We then developed high resolution maps of the disease's estimated distribution using a boosted regression tree approach. Models were developed using nine climatic and environmental covariates that are related to the MAYV transmission cycle. Using the output of our boosted regression tree models, we estimated the total population living in regions suitable for MAYV transmission. The evidence consensus scores revealed high or very high evidence of MAYV transmission in several countries including Brazil (especially the states of Mato Grosso and Goiás), Venezuela, Peru, Trinidad and Tobago, and French Guiana. According to the boosted regression tree models, a substantial region of South America is suitable for MAYV transmission, including north and central Brazil, French Guiana, and Suriname. Some regions (e.g., Guyana) with only moderate evidence of known transmission were identified as highly suitable for MAYV. We estimate that approximately 58.9 million people (95% CI: 21.4-100.4) in Central and South America live in areas that may be suitable for MAYV transmission, including 46.2 million people (95% CI: 17.6-68.9) in Brazil. Our results may assist in prioritizing high-risk areas for vector control, human disease surveillance and ecological studies.
Collapse
Affiliation(s)
- Michael Celone
- Department of Preventive Medicine & Biostatistics, Uniformed Services University of the Health Sciences, F. Edward Hébert School of Medicine, Bethesda, Maryland, United States of America
| | - Sean Beeman
- Department of Preventive Medicine & Biostatistics, Uniformed Services University of the Health Sciences, F. Edward Hébert School of Medicine, Bethesda, Maryland, United States of America
| | - Barbara A. Han
- Cary Institute of Ecosystem Studies, Millbrook, New York, United States of America
| | - Alexander M. Potter
- One Health Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Walter Reed Biosystematics Unit, Smithsonian Museum Support Center, Suitland, Maryland, United States of America
- Department of Entomology, Smithsonian Institution—National Museum of Natural History (NMNH), Washington, DC, United States of America
| | - David B. Pecor
- One Health Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Walter Reed Biosystematics Unit, Smithsonian Museum Support Center, Suitland, Maryland, United States of America
- Department of Entomology, Smithsonian Institution—National Museum of Natural History (NMNH), Washington, DC, United States of America
| | - Bernard Okech
- Department of Preventive Medicine & Biostatistics, Uniformed Services University of the Health Sciences, F. Edward Hébert School of Medicine, Bethesda, Maryland, United States of America
| | - Simon Pollett
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| |
Collapse
|
43
|
Côrtes N, Lira A, Prates-Syed W, Dinis Silva J, Vuitika L, Cabral-Miranda W, Durães-Carvalho R, Balan A, Cabral-Marques O, Cabral-Miranda G. Integrated control strategies for dengue, Zika, and Chikungunya virus infections. Front Immunol 2023; 14:1281667. [PMID: 38196945 PMCID: PMC10775689 DOI: 10.3389/fimmu.2023.1281667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/24/2023] [Indexed: 01/11/2024] Open
Abstract
Arboviruses are a major threat to public health in tropical regions, encompassing over 534 distinct species, with 134 capable of causing diseases in humans. These viruses are transmitted through arthropod vectors that cause symptoms such as fever, headache, joint pains, and rash, in addition to more serious cases that can lead to death. Among the arboviruses, dengue virus stands out as the most prevalent, annually affecting approximately 16.2 million individuals solely in the Americas. Furthermore, the re-emergence of the Zika virus and the recurrent outbreaks of chikungunya in Africa, Asia, Europe, and the Americas, with one million cases reported annually, underscore the urgency of addressing this public health challenge. In this manuscript we discuss the epidemiology, viral structure, pathogenicity and integrated control strategies to combat arboviruses, and the most used tools, such as vaccines, monoclonal antibodies, treatment, etc., in addition to presenting future perspectives for the control of arboviruses. Currently, specific medications for treating arbovirus infections are lacking, and symptom management remains the primary approach. However, promising advancements have been made in certain treatments, such as Chloroquine, Niclosamide, and Isatin derivatives, which have demonstrated notable antiviral properties against these arboviruses in vitro and in vivo experiments. Additionally, various strategies within vector control approaches have shown significant promise in reducing arbovirus transmission rates. These encompass public education initiatives, targeted insecticide applications, and innovative approaches like manipulating mosquito bacterial symbionts, such as Wolbachia. In conclusion, combatting the global threat of arbovirus diseases needs a comprehensive approach integrating antiviral research, vaccination, and vector control. The continued efforts of research communities, alongside collaborative partnerships with public health authorities, are imperative to effectively address and mitigate the impact of these arboviral infections on public health worldwide.
Collapse
Affiliation(s)
- Nelson Côrtes
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- The Interunits Graduate Program in Biotechnology of the University of São Paulo, the Butantan Institute and the Technological Research Institute of the State of São Paulo, São Paulo, Brazil
| | - Aline Lira
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- The Interunits Graduate Program in Biotechnology of the University of São Paulo, the Butantan Institute and the Technological Research Institute of the State of São Paulo, São Paulo, Brazil
| | - Wasim Prates-Syed
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- The Interunits Graduate Program in Biotechnology of the University of São Paulo, the Butantan Institute and the Technological Research Institute of the State of São Paulo, São Paulo, Brazil
| | - Jaqueline Dinis Silva
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- The Graduate Program in Pathophysiology and Toxicology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Larissa Vuitika
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Ricardo Durães-Carvalho
- São Paulo School of Medicine, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Andrea Balan
- The Interunits Graduate Program in Biotechnology of the University of São Paulo, the Butantan Institute and the Technological Research Institute of the State of São Paulo, São Paulo, Brazil
- Applied Structural Biology Laboratory, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Otavio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- The Graduate Program in Pathophysiology and Toxicology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Medicine, Division of Molecular Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Gustavo Cabral-Miranda
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- The Interunits Graduate Program in Biotechnology of the University of São Paulo, the Butantan Institute and the Technological Research Institute of the State of São Paulo, São Paulo, Brazil
- The Graduate Program in Pathophysiology and Toxicology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
44
|
Adams LJ, Raju S, Ma H, Gilliland T, Reed DS, Klimstra WB, Fremont DH, Diamond MS. Structural and functional basis of VLDLR receptor usage by Eastern equine encephalitis virus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.15.567188. [PMID: 38014196 PMCID: PMC10680733 DOI: 10.1101/2023.11.15.567188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The very low-density lipoprotein receptor (VLDLR) is comprised of eight LDLR type A (LA) domains and supports entry of distantly related Eastern equine encephalitis (EEEV) and Semliki Forest (SFV) alphaviruses. Here, by resolving multiple cryo-electron microscopy structures of EEEV-VLDLR complexes and performing mutagenesis and functional studies, we show that EEEV uses multiple sites (E1/E2 cleft and E2 A domain) to engage different LA domains simultaneously. However, no single LA domain is necessary or sufficient to support efficient EEEV infection, highlighting complexity in domain usage. Whereas all EEEV strains show conservation of two VLDLR binding sites, the EEEV PE-6 strain and other EEE complex members feature a single amino acid substitution that mediates binding of LA domains to an additional site on the E2 B domain. These structural and functional analyses informed the design of a minimal VLDLR decoy receptor that neutralizes EEEV infection and protects mice from lethal challenge.
Collapse
Affiliation(s)
- Lucas J. Adams
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Saravanan Raju
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hongming Ma
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Theron Gilliland
- The Center for Vaccine Research and Department of Immunology, The University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Douglas S. Reed
- The Center for Vaccine Research and Department of Immunology, The University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - William B. Klimstra
- The Center for Vaccine Research and Department of Immunology, The University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Daved H. Fremont
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael S. Diamond
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, Saint Louis, MO 63110, USA
| |
Collapse
|
45
|
Periferakis A, Periferakis AT, Troumpata L, Periferakis K, Scheau AE, Savulescu-Fiedler I, Caruntu A, Badarau IA, Caruntu C, Scheau C. Kaempferol: A Review of Current Evidence of Its Antiviral Potential. Int J Mol Sci 2023; 24:16299. [PMID: 38003488 PMCID: PMC10671393 DOI: 10.3390/ijms242216299] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/07/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023] Open
Abstract
Kaempferol and its derivatives are flavonoids found in various plants, and a considerable number of these have been used in various medical applications worldwide. Kaempferol and its compounds have well-known antioxidant, anti-inflammatory and antimicrobial properties among other health benefits. However, the antiviral properties of kaempferol are notable, and there is a significant number of experimental studies on this topic. Kaempferol compounds were effective against DNA viruses such as hepatitis B virus, viruses of the alphaherpesvirinae family, African swine fever virus, and pseudorabies virus; they were also effective against RNA viruses, namely feline SARS coronavirus, dengue fever virus, Japanese encephalitis virus, influenza virus, enterovirus 71, poliovirus, respiratory syncytial virus, human immunodeficiency virus, calicivirus, and chikungunya virus. On the other hand, no effectiveness against murine norovirus and hepatitis A virus could be determined. The antiviral action mechanisms of kaempferol compounds are various, such as the inhibition of viral polymerases and of viral attachment and entry into host cells. Future research should be focused on further elucidating the antiviral properties of kaempferol compounds from different plants and assessing their potential use to complement the action of antiviral drugs.
Collapse
Affiliation(s)
- Argyrios Periferakis
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Akadimia of Ancient Greek and Traditional Chinese Medicine, 16675 Athens, Greece
- Elkyda, Research & Education Centre of Charismatheia, 17675 Athens, Greece
| | - Aristodemos-Theodoros Periferakis
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Elkyda, Research & Education Centre of Charismatheia, 17675 Athens, Greece
| | - Lamprini Troumpata
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Konstantinos Periferakis
- Akadimia of Ancient Greek and Traditional Chinese Medicine, 16675 Athens, Greece
- Pan-Hellenic Organization of Educational Programs (P.O.E.P), 17236 Athens, Greece
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Ioana Anca Badarau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
| |
Collapse
|
46
|
Payet M, Septembre-Malaterre A, Gasque P, Guillot X. Human Synovial Mesenchymal Stem Cells Expressed Immunoregulatory Factors IDO and TSG6 in a Context of Arthritis Mediated by Alphaviruses. Int J Mol Sci 2023; 24:15932. [PMID: 37958918 PMCID: PMC10649115 DOI: 10.3390/ijms242115932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/26/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Infection by arthritogenic alphaviruses (aavs) can lead to reactive arthritis, which is characterized by inflammation and persistence of the virus; however, its mechanisms remain ill-characterized. Intriguingly, it has been shown that viral persistence still takes place in spite of robust innate and adaptive immune responses, characterized notably by the infiltration of macrophages (sources of TNF-alpha) as well as T/NK cells (sources of IFN-gamma) in the infected joint. Aavs are known to target mesenchymal stem cells (MSCs) in the synovium, and we herein tested the hypothesis that the infection of MSCs may promote the expression of immunoregulators to skew the anti-viral cellular immune responses. We compared the regulated expression via human synovial MSCs of pro-inflammatory mediators (e.g., IL-1β, IL6, CCL2, miR-221-3p) to that of immunoregulators (e.g., IDO, TSG6, GAS6, miR146a-5p). We used human synovial tissue-derived MSCs which were infected with O'Nyong-Nyong alphavirus (ONNV, class II aav) alone, or combined with recombinant human TNF-α or IFN-γ, to mimic the clinical settings. We confirmed via qPCR and immunofluorescence that ONNV infected human synovial tissue-derived MSCs. Interestingly, ONNV alone did not regulate the expression of pro-inflammatory mediators. In contrast, IDO, TSG6, and GAS6 mRNA expression were increased in response to ONNV infection alone, but particularly when combined with both recombinant cytokines. ONNV infection equally decreased miR-146a-5p and miR-221-3p in the untreated cells and abrogated the stimulatory activity of the recombinant TNF-α but not the IFN-gamma. Our study argues for a major immunoregulatory phenotype of MSCs infected with ONNV which may favor virus persistence in the inflamed joint.
Collapse
Affiliation(s)
- Melissa Payet
- Research Unit ‘Etudes Pharmaco-Immunologiques’ UR EPI, Université de la Réunion, 97400 Saint-Denis, La Réunion, France; (M.P.); (A.S.-M.)
| | - Axelle Septembre-Malaterre
- Research Unit ‘Etudes Pharmaco-Immunologiques’ UR EPI, Université de la Réunion, 97400 Saint-Denis, La Réunion, France; (M.P.); (A.S.-M.)
| | - Philippe Gasque
- Research Unit ‘Etudes Pharmaco-Immunologiques’ UR EPI, Université de la Réunion, 97400 Saint-Denis, La Réunion, France; (M.P.); (A.S.-M.)
- Immunology Laboratory (LICE-OI), CHU Bellepierre, Reunion University Hospital, 97400 Saint-Denis, La Réunion, France
| | - Xavier Guillot
- Research Unit ‘Etudes Pharmaco-Immunologiques’ UR EPI, Université de la Réunion, 97400 Saint-Denis, La Réunion, France; (M.P.); (A.S.-M.)
- Rheumatology Clinical Department, CHU Bellepierre, Reunion University Hospital, 97400 Saint-Denis, La Réunion, France
| |
Collapse
|
47
|
Zhang W, Wang J, Liu Q, Gong Z. A Review of Pathogens Transmitted by the Container-Inhabiting Mosquitoes, Aedes Albopictus, A Global Public Health Threat. China CDC Wkly 2023; 5:984-990. [PMID: 38023389 PMCID: PMC10652089 DOI: 10.46234/ccdcw2023.185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Dengue virus (DENV), Chikungunya virus (CHIKV), and Zika virus (ZIKV) are highly pathogenic human arboviruses transmitted by the Aedes (Stegomyia) albopictus (Skuse) (Diptera: Culicidae) or Ae. Albopictus mosquito. These arboviruses are responsible for causing fever, hemorrhagic conditions, and neurological diseases in humans post-bite from an infected Aedes mosquito. Over the past 80 years, the Ae. albopictus has infested every habitable continent, bar Antarctica, thereby escalating the probability of global insect-borne infectious disease outbreaks. This research follows the global transmission pattern of Ae. albopictus and provides a summary of disease prevention and control strategies for mosquito-borne infections, as implemented by the World Health Organization (WHO) and both Asian and European countries. Consequently, this study can aid in the prevention and control of mosquito-borne diseases while acting as a basis for international collaboration on effectively managing arbovirus infection issues in public health.
Collapse
Affiliation(s)
- Wenrong Zhang
- School of Public Health, Hangzhou Medical College, Hangzhou City, Zhejiang Province, China
| | - Jinna Wang
- Institute of Infectious Disease Prevention and Control, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou City, Zhejiang Province, China
| | - Qinmei Liu
- Institute of Infectious Disease Prevention and Control, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou City, Zhejiang Province, China
| | - Zhenyu Gong
- Institute of Infectious Disease Prevention and Control, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou City, Zhejiang Province, China
| |
Collapse
|
48
|
Manzato VDM, Di Santo C, Torquato RJS, Coelho C, Gallo G, Hardy L, Würtele M, Tanaka AS. Boophilin D1, a Kunitz type protease inhibitor, as a source of inhibitors for the ZIKA virus NS2B-NS3 protease. Biochimie 2023; 214:96-101. [PMID: 37364769 DOI: 10.1016/j.biochi.2023.06.010] [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: 01/17/2023] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 06/28/2023]
Abstract
Arboviruses are a global concern for a multitude of reasons, including their increased incidence and human mortality. Vectors associated with arboviruses include the mosquito Aedes sp., which is responsible for transmitting the Zika virus. Flaviviruses, like the Zika virus, present only one chymotrypsin-like serine protease (NS3) in their genome. Together with host enzymes, the NS2B co-factor NS3 protease complex are essential for the viral replication cycle by virus polyprotein processing. To search for Zika virus NS2B-NS3 protease (ZIKVPro) inhibitors, a phage display library was constructed using the Boophilin domain 1 (BoophD1), a thrombin inhibitor from the Kunitz family. A BoophilinD1 library mutated at positions P1-P4' was constructed, presenting a titer of 2.9x106 (cfu), and screened utilizing purified ZIKVPro. The results demonstrated at the P1-P4' positions the occurrence of 47% RALHA sequence (mut 12) and 11.8% RASWA sequence (mut14), SMRPT, or KALIP (wt) sequence. BoophD1-wt and mutants 12 and 14 were expressed and purified. The purified BoophD1 wt, mut 12 and 14, presented Ki values for ZIKVPro of 0.103, 0.116, and 0.101 μM, respectively. The BoophD1 mutant inhibitors inhibit the Dengue virus 2 protease (DENV2) with Ki values of 0.298, 0.271, and 0.379 μM, respectively. In conclusion, BoophD1 mut 12 and 14 selected for ZIKVPro demonstrated inhibitory activity like BoophD1-wt, suggesting that these are the strongest Zika inhibitors present in the BoophD1 mutated phage display library. Furthermore, BoophD1 mutants selected for ZIKVPro inhibit both Zika and Dengue 2 proteases making them potential pan-flavivirus inhibitors.
Collapse
Affiliation(s)
- Veronica de Moraes Manzato
- Department of Biochemistry, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Camila Di Santo
- Department of Biochemistry, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Ricardo Jose Soares Torquato
- Department of Biochemistry, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Camila Coelho
- Department of Science and Technology, Federal University of São Paulo (UNIFESP), São José dos Campos, SP, Brazil
| | - Gloria Gallo
- Department of Science and Technology, Federal University of São Paulo (UNIFESP), São José dos Campos, SP, Brazil
| | - Leon Hardy
- Department of Physics, University of South Florida, Tampa, USA
| | - Martin Würtele
- Department of Science and Technology, Federal University of São Paulo (UNIFESP), São José dos Campos, SP, Brazil
| | - Aparecida Sadae Tanaka
- Department of Biochemistry, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), RJ, Brazil.
| |
Collapse
|
49
|
Powers AM, Williamson LE, Carnahan RH, Crowe JE, Hyde JL, Jonsson CB, Nasar F, Weaver SC. Developing a Prototype Pathogen Plan and Research Priorities for the Alphaviruses. J Infect Dis 2023; 228:S414-S426. [PMID: 37849399 PMCID: PMC11007399 DOI: 10.1093/infdis/jiac326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023] Open
Abstract
The Togaviridae family, genus, Alphavirus, includes several mosquito-borne human pathogens with the potential to spread to near pandemic proportions. Most of these are zoonotic, with spillover infections of humans and domestic animals, but a few such as chikungunya virus (CHIKV) have the ability to use humans as amplification hosts for transmission in urban settings and explosive outbreaks. Most alphaviruses cause nonspecific acute febrile illness, with pathogenesis sometimes leading to either encephalitis or arthralgic manifestations with severe and chronic morbidity and occasional mortality. The development of countermeasures, especially against CHIKV and Venezuelan equine encephalitis virus that are major threats, has included vaccines and antibody-based therapeutics that are likely to also be successful for rapid responses with other members of the family. However, further work with these prototypes and other alphavirus pathogens should target better understanding of human tropism and pathogenesis, more comprehensive identification of cellular receptors and entry, and better understanding of structural mechanisms of neutralization.
Collapse
Affiliation(s)
- Ann M Powers
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, USA
| | - Lauren E Williamson
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Robert H Carnahan
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James E Crowe
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, USA
| | - Jennifer L Hyde
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Colleen B Jonsson
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Farooq Nasar
- Emerging Infectious Diseases Branch and Viral Disease Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Scott C Weaver
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
- World Reference Center for Emerging Viruses and Arboviruses, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
50
|
Lata K, Charles S, Mangala Prasad V. Advances in computational approaches to structure determination of alphaviruses and flaviviruses using cryo-electron microscopy. J Struct Biol 2023; 215:107993. [PMID: 37414374 DOI: 10.1016/j.jsb.2023.107993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/15/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
Advancements in the field of cryo-electron microscopy (cryo-EM) have greatly contributed to our current understanding of virus structures and life cycles. In this review, we discuss the application of single particle cryo-electron microscopy (EM) for the structure elucidation of small enveloped icosahedral viruses, namely, alpha- and flaviviruses. We focus on technical advances in cryo-EM data collection, image processing, three-dimensional reconstruction, and refinement strategies for obtaining high-resolution structures of these viruses. Each of these developments enabled new insights into the alpha- and flavivirus architecture, leading to a better understanding of their biology, pathogenesis, immune response, immunogen design, and therapeutic development.
Collapse
Affiliation(s)
- Kiran Lata
- Molecular Biophysics Unit, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Sylvia Charles
- Molecular Biophysics Unit, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Vidya Mangala Prasad
- Molecular Biophysics Unit, Indian Institute of Science, Bengaluru, Karnataka 560012, India; Center for Infectious Disease Research, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| |
Collapse
|