1
|
Yuya W, Yuansong Y, Susu L, Chen L, Yong W, Yining W, YouChun W, Changfa F. Progress and challenges in development of animal models for dengue virus infection. Emerg Microbes Infect 2024; 13:2404159. [PMID: 39312399 PMCID: PMC11423536 DOI: 10.1080/22221751.2024.2404159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/14/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024]
Abstract
ABSTRACTThe severity of the dengue epidemic is on the rise, with its geographic range had expanded to southern Europe by 2024. In this August, the WHO updated the pathogens that could spark the next pandemic, dengue virus was on the list. Vaccines and drugs serve as powerful tools for both preventing dengue infections and treating patients. Animal models play a pivotal role in vaccine development and drug screening. Available potential susceptible animals, including non-human primates, rodents, pigs, and tree shrews, have been extensively explored to establish animal models of dengue disease. Despite significant advancements, there are still notable limitations. Different animal models exhibit distinct constraining factors such as viraemia, host susceptibility, immune function of the host, clinical symptoms, ADE (antibody-dependent enhancement) phenomena, cytokine storm response to various serotypes and strain variations. Furthermore, despite extensive research on the dengue virus receptor in recent years, genetically modified animal models immunocompetent harbouring dengue virus susceptibility receptors have not yet been available. This work reviewed the research progress of dengue virus receptors and dengue animal models, suggesting that the development of genetically modified murine models expressing dengue virus functional receptors may hold a promise for future dengue disease research, especially for its vaccine development.
Collapse
Affiliation(s)
- Wang Yuya
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Yang Yuansong
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Liu Susu
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Ling Chen
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
- College of Life Science school, Northwest University, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Xi’an, People’s Republic of China
| | - Wu Yong
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Wang Yining
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Wang YouChun
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, People’s Republic of China
| | - Fan Changfa
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| |
Collapse
|
2
|
Edgar JE, Bournazos S. Fc-FcγR interactions during infections: From neutralizing antibodies to antibody-dependent enhancement. Immunol Rev 2024. [PMID: 39268652 DOI: 10.1111/imr.13393] [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] [Indexed: 09/17/2024]
Abstract
Advances in antibody technologies have resulted in the development of potent antibody-based therapeutics with proven clinical efficacy against infectious diseases. Several monoclonal antibodies (mAbs), mainly against viruses such as SARS-CoV-2, HIV-1, Ebola virus, influenza virus, and hepatitis B virus, are currently undergoing clinical testing or are already in use. Although these mAbs exhibit potent neutralizing activity that effectively blocks host cell infection, their antiviral activity results not only from Fab-mediated virus neutralization, but also from the protective effector functions mediated through the interaction of their Fc domains with Fcγ receptors (FcγRs) on effector leukocytes. Fc-FcγR interactions confer pleiotropic protective activities, including the clearance of opsonized virions and infected cells, as well as the induction of antiviral T-cell responses. However, excessive or inappropriate activation of specific FcγR pathways can lead to disease enhancement and exacerbated pathology, as seen in the context of dengue virus infections. A comprehensive understanding of the diversity of Fc effector functions during infection has guided the development of engineered antiviral antibodies optimized for maximal effector activity, as well as the design of targeted therapeutic approaches to prevent antibody-dependent enhancement of disease.
Collapse
Affiliation(s)
- Julia E Edgar
- The London School of Hygiene and Tropical Medicine, London, UK
| | - Stylianos Bournazos
- The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, New York, USA
| |
Collapse
|
3
|
Gupta A, Kao KS, Yamin R, Oren DA, Goldgur Y, Du J, Lollar P, Sundberg EJ, Ravetch JV. Mechanism of glycoform specificity and in vivo protection by an anti-afucosylated IgG nanobody. Nat Commun 2023; 14:2853. [PMID: 37202422 PMCID: PMC10195009 DOI: 10.1038/s41467-023-38453-1] [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: 02/03/2023] [Accepted: 05/03/2023] [Indexed: 05/20/2023] Open
Abstract
Immunoglobulin G (IgG) antibodies contain a complex N-glycan embedded in the hydrophobic pocket between its heavy chain protomers. This glycan contributes to the structural organization of the Fc domain and determines its specificity for Fcγ receptors, thereby dictating distinct cellular responses. The variable construction of this glycan structure leads to highly-related, but non-equivalent glycoproteins known as glycoforms. We previously reported synthetic nanobodies that distinguish IgG glycoforms. Here, we present the structure of one such nanobody, X0, in complex with the Fc fragment of afucosylated IgG1. Upon binding, the elongated CDR3 loop of X0 undergoes a conformational shift to access the buried N-glycan and acts as a 'glycan sensor', forming hydrogen bonds with the afucosylated IgG N-glycan that would otherwise be sterically hindered by the presence of a core fucose residue. Based on this structure, we designed X0 fusion constructs that disrupt pathogenic afucosylated IgG1-FcγRIIIa interactions and rescue mice in a model of dengue virus infection.
Collapse
Affiliation(s)
- Aaron Gupta
- Laboratory of Molecular Genetics & Immunology, The Rockefeller University, New York, NY, USA
| | - Kevin S Kao
- Laboratory of Molecular Genetics & Immunology, The Rockefeller University, New York, NY, USA
| | - Rachel Yamin
- Laboratory of Molecular Genetics & Immunology, The Rockefeller University, New York, NY, USA
| | - Deena A Oren
- Structural Biology Resource Center, The Rockefeller University, New York, NY, USA
| | - Yehuda Goldgur
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonathan Du
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Pete Lollar
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric J Sundberg
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Jeffrey V Ravetch
- Laboratory of Molecular Genetics & Immunology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
4
|
Wilken L, Stelz S, Prajeeth CK, Rimmelzwaan GF. Transient Blockade of Type I Interferon Signalling Promotes Replication of Dengue Virus Strain D2Y98P in Adult Wild-Type Mice. Viruses 2023; 15:v15040814. [PMID: 37112795 PMCID: PMC10142689 DOI: 10.3390/v15040814] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 04/29/2023] Open
Abstract
Dengue virus serotypes 1 to 4 (DENV1-4) place nearly half the global population at risk of infection and the licenced tetravalent dengue vaccine fails to protect individuals who have not previously been exposed to DENV. The development of intervention strategies had long been hampered by the lack of a suitable small animal model. DENV does not replicate in wild-type mice due to its inability to antagonise the mouse type I interferon (IFN) response. Mice deficient in type I IFN signalling (Ifnar1-/- mice) are highly susceptible to DENV infection, but their immunocompromised status makes it difficult to interpret immune responses elicited by experimental vaccines. To develop an alternative mouse model for vaccine testing, we treated adult wild-type mice with MAR1-5A3-an IFNAR1-blocking, non-cell-depleting antibody-prior to infection with the DENV2 strain D2Y98P. This approach would allow for vaccination of immunocompetent mice and subsequent inhibition of type I IFN signalling prior to challenge infection. While Ifnar1-/- mice quickly succumbed to infection, MAR1-5A3-treated mice did not show any signs of illness but eventually seroconverted. Infectious virus was recovered from the sera and visceral organs of Ifnar1-/- mice, but not from those of mice treated with MAR1-5A3. However, high levels of viral RNA were detected in the samples of MAR1-5A3-treated mice, indicating productive viral replication and dissemination. This transiently immunocompromised mouse model of DENV2 infection will aid the pre-clinical assessment of next-generation vaccines as well as novel antiviral treatments.
Collapse
Affiliation(s)
- Lucas Wilken
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), 30559 Hannover, Germany
| | - Sonja Stelz
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), 30559 Hannover, Germany
| | - Chittappen Kandiyil Prajeeth
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), 30559 Hannover, Germany
| | - Guus F Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), 30559 Hannover, Germany
| |
Collapse
|
5
|
Gupta A, Kao K, Yamin R, Oren DA, Goldgur Y, Du J, Lollar P, Sundberg EJ, Ravetch JV. Mechanism of glycoform specificity and protection against antibody dependent enhancement by an anti-afucosylated IgG nanobody. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.23.525277. [PMID: 36747840 PMCID: PMC9900767 DOI: 10.1101/2023.01.23.525277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Immunoglobulin G (IgG) antibodies contain a single, complex N -glycan on each IgG heavy chain protomer embedded in the hydrophobic pocket between its Cγ2 domains. The presence of this glycan contributes to the structural organization of the Fc domain and determines its specificity for Fcγ receptors, thereby determining distinct cellular responses. On the Fc, the variable construction of this glycan structure leads to a family of highly-related, but non-equivalent glycoproteins known as glycoforms. We previously reported the development of synthetic nanobodies that distinguish IgG glycoforms without cross-reactivity to off-target glycoproteins or free glycans. Here, we present the X-ray crystal structure of one such nanobody, X0, in complex with its specific binding partner, the Fc fragment of afucosylated IgG1. Two X0 nanobodies bind a single afucosylated Fc homodimer at the upper Cγ2 domain, making both protein-protein and protein-carbohydrate contacts and overlapping the binding site for Fcγ receptors. Upon binding, the elongated CDR3 loop of X0 undergoes a conformational shift to access the buried N -glycan and acts as a 'glycan sensor', forming hydrogen bonds with the afucosylated IgG N -glycan that would otherwise be sterically hindered by the presence of a core fucose residue. Based on this structure, we designed X0 fusion constructs that disrupt pathogenic afucosylated IgG1-FcγRIIIa interactions and rescue mice in a model of dengue virus infection.
Collapse
|
6
|
Shukla R, Ahuja R, Beesetti H, Garg A, Aggarwal C, Chaturvedi S, Nayyar K, Arora U, Lal AA, Khanna N. Sinococuline, a bioactive compound of Cocculus hirsutus has potent anti-dengue activity. Sci Rep 2023; 13:1026. [PMID: 36658277 PMCID: PMC9852271 DOI: 10.1038/s41598-023-27927-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/10/2023] [Indexed: 01/20/2023] Open
Abstract
Dengue virus (DENV) infection has increased worldwide, with over 400 million infections annually, and has become a serious public health concern. Several drug candidates, new and repurposed, have failed to meet the primary efficacy endpoints. We have recently shown that Aqueous Extract of the stem of Cocculus hirsutus (AQCH) was effective in vitro and in vivo against DENV and was safe in humans. We now report that an active ingredient of AQCH, Sinococuline, protects against the antibody-mediated secondary-DENV infection in the AG129 mouse model. DENV infection markers were assessed, viz. serum viremia and vital organs pathologies-viral load, proinflammatory cytokines and intestinal vascular leakage. The treatment with Sinococuline at 2.0 mg/kg/day; BID (twice a day), was the most effective in protecting the severely DENV-infected AG129 mice. Also, this dose effectively reduced serum viremia and tissue-viral load and inhibited the elevated expression levels of proinflammatory cytokines (TNF-α and IL-6) in several vital organs. Based on these findings, it could be explored further for pre-clinical and clinical developments for the treatment of dengue.
Collapse
Affiliation(s)
- Rahul Shukla
- Translational Health, Molecular Medicine Division, International Centre for Genetic Engineering and Biotechnology, New Delhi, India.,Division of Virus Research and Therapeutics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Richa Ahuja
- Translational Health, Molecular Medicine Division, International Centre for Genetic Engineering and Biotechnology, New Delhi, India.,Sun Pharmaceutical Industries Limited, Gurugram, India
| | - Hemalatha Beesetti
- Sun Pharmaceutical Industries Limited, Gurugram, India.,Virology Division, Foundation for Neglected Disease Research, 20A, KIADB Industrial Area Veerapura, Doddaballapur, Bengaluru, Karnataka, 561203, India
| | - Amit Garg
- Translational Health, Molecular Medicine Division, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Charu Aggarwal
- Translational Health, Molecular Medicine Division, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Shivam Chaturvedi
- Translational Health, Molecular Medicine Division, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | | | - Upasana Arora
- Sun Pharmaceutical Industries Limited, Gurugram, India
| | - Altaf A Lal
- Sun Pharmaceutical Industries Limited, Gurugram, India
| | - Navin Khanna
- Translational Health, Molecular Medicine Division, International Centre for Genetic Engineering and Biotechnology, New Delhi, India. .,Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, India.
| |
Collapse
|
7
|
Yan Q, Zheng W, Jiang Y, Zhou P, Lai Y, Liu C, Wu P, Zhuang H, Huang H, Li G, Zhan S, Lao Z, Liu X. Transcriptomic reveals the ferroptosis features of host response in a mouse model of Zika virus infection. J Med Virol 2023; 95:e28386. [PMID: 36477858 DOI: 10.1002/jmv.28386] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 11/15/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022]
Abstract
Zika virus (ZIKV) is a neurotropic flavivirus. The outbreak of ZIKV in 2016 created a global health emergency. However, the underlying pathogenic mechanisms remain elusive. We investigated the host response features of in vivo replication in a mouse model of ZIKV infection, by performing a series of transcriptomic and bioinformatic analyses of ZIKV and mock-infected brain tissue. Tissue damage, inflammatory cells infiltration and high viral replication were observed in the brain tissue of ZIKV infected mice. RNA-Seq of the brain indicated the activation of ferroptosis pathways. Enrichment analysis of ferroptosis regulators revealed their involvement in pathways such as mineral absorption, fatty acid biosynthesis, fatty acid degradation, PPAR signaling pathway, peroxidase, and adipokinesine signalling pathway. We then identified 12 interacted hub ferroptosis regulators (CYBB, HMOX1, CP, SAT1, TF, SLC39A14, FTL, LPCAT3, FTH1, SLC3A2, TP53, and SLC40A1) that were related to the differential expression of CD8+ T cells, microglia and monocytes. CYBB, HMOX1, SALT, and SLAC40A1 were selected as potential biomarkers of ZIKV infection. Finally, we validated our results using RT-qPCR and outside available datasets. For the first time, we proposed a possible mechanism of ferroptosis in brain tissue infected by ZIKV in mice and identified the four key ferroptosis regulators.
Collapse
Affiliation(s)
- Qian Yan
- Department of Internal Medicine of Traditional Chinese Medicine (TCM), The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Pulmonary and Critical Care Medicine (PCCM), The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.,Animal Biosafety Level 2 laboratory (ABSL-2), Animal Laboratory Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenjiang Zheng
- Department of Internal Medicine of Traditional Chinese Medicine (TCM), The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Pulmonary and Critical Care Medicine (PCCM), The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.,Animal Biosafety Level 2 laboratory (ABSL-2), Animal Laboratory Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong Jiang
- Traditional Chinese Medicine Innovation Research Center and Department of Respiratory Medicine, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China
| | - Peiwen Zhou
- Animal Biosafety Level 3 laboratory (ABSL-3), Foshan Institute of Medical Microbiology, Foshan, China
| | - Yanni Lai
- Department of Diagnostics of Traditional Chinese Medicine (TCM), Basic Medical Sciences School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chengxin Liu
- Department of Internal Medicine of Traditional Chinese Medicine (TCM), The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peng Wu
- Department of Internal Medicine of Traditional Chinese Medicine (TCM), The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongfa Zhuang
- Department of Pulmonary and Critical Care Medicine (PCCM), The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huiting Huang
- Department of Pulmonary and Critical Care Medicine (PCCM), The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Geng Li
- Animal Biosafety Level 2 laboratory (ABSL-2), Animal Laboratory Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shaofeng Zhan
- Department of Internal Medicine of Traditional Chinese Medicine (TCM), The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Pulmonary and Critical Care Medicine (PCCM), The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zizhao Lao
- Animal Biosafety Level 2 laboratory (ABSL-2), Animal Laboratory Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Traditional Chinese Medicine Innovation Research Center and Department of Respiratory Medicine, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China
| | - Xiaohong Liu
- Department of Internal Medicine of Traditional Chinese Medicine (TCM), The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Pulmonary and Critical Care Medicine (PCCM), The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
8
|
Chiang CY, Chen MY, Hsu CW, Liu CY, Tsai YW, Liao HC, Yan JY, Chuang ZS, Wang HI, Pan CH, Yu CY, Yu GY, Liao CL, Liu SJ, Chen HW. Induction of high affinity monoclonal antibodies against SARS-CoV-2 variant infection using a DNA prime-protein boost strategy. J Biomed Sci 2022; 29:37. [PMID: 35681239 PMCID: PMC9178533 DOI: 10.1186/s12929-022-00823-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 06/03/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Calls for the coronavirus to be treated as an endemic illness, such as the flu, are increasing. After achieving high coverage of COVID-19 vaccination, therapeutic drugs have become important for future SARS-CoV-2 variant outbreaks. Although many monoclonal antibodies have been approved for emergency use as treatments for SARS-CoV-2 infection, some monoclonal antibodies are not authorized for variant treatment. Broad-spectrum monoclonal antibodies are unmet medical needs. METHODS We used a DNA prime-protein boost approach to generate high-quality monoclonal antibodies. A standard ELISA was employed for the primary screen, and spike protein-human angiotensin-converting enzyme 2 blocking assays were used for the secondary screen. The top 5 blocking clones were selected for further characterization, including binding ability, neutralization potency, and epitope mapping. The therapeutic effects of the best monoclonal antibody against SARS-CoV-2 infection were evaluated in a hamster infection model. RESULTS Several monoclonal antibodies were selected that neutralize different SARS-CoV-2 variants of concern (VOCs). These VOCs include Alpha, Beta, Gamma, Delta, Kappa and Lambda variants. The high neutralizing antibody titers against the Beta variant would be important to treat Beta-like variants. Among these monoclonal antibodies, mAb-S5 displays the best potency in terms of binding affinity and neutralizing capacity. Importantly, mAb-S5 protects animals from SARS-CoV-2 challenge, including the Wuhan strain, D614G, Alpha and Delta variants, although mAb-S5 exhibits decreased neutralization potency against the Delta variant. Furthermore, the identified neutralizing epitopes of monoclonal antibodies are all located in the receptor-binding domain (RBD) of the spike protein but in different regions. CONCLUSIONS Our approach generates high-potency monoclonal antibodies against a broad spectrum of VOCs. Multiple monoclonal antibody combinations may be the best strategy to treat future SARS-CoV-2 variant outbreaks.
Collapse
Affiliation(s)
- Chen-Yi Chiang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Mei-Yu Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Chia-Wei Hsu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Chia-Yeh Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Yu-Wen Tsai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Hung-Chun Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 35053, Taiwan
- Department of Life Sciences, National Tsing Hua University, Hsinchu, 30072, Taiwan
| | - Jia-Ying Yan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Zih-Shiuan Chuang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Hsin-I Wang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Chien-Hsiung Pan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Chia-Yi Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Guann-Yi Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Ching-Len Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Shih-Jen Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 35053, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan.
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, 307378, Taiwan.
| | - Hsin-Wei Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, 35053, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan.
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, 307378, Taiwan.
| |
Collapse
|
9
|
Farouq MAH, Acevedo R, Ferro VA, Mulheran PA, Al Qaraghuli MM. The Role of Antibodies in the Treatment of SARS-CoV-2 Virus Infection, and Evaluating Their Contribution to Antibody-Dependent Enhancement of Infection. Int J Mol Sci 2022; 23:6078. [PMID: 35682757 PMCID: PMC9181534 DOI: 10.3390/ijms23116078] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/26/2022] [Accepted: 05/26/2022] [Indexed: 02/06/2023] Open
Abstract
Antibodies play a crucial role in the immune response, in fighting off pathogens as well as helping create strong immunological memory. Antibody-dependent enhancement (ADE) occurs when non-neutralising antibodies recognise and bind to a pathogen, but are unable to prevent infection, and is widely known and is reported as occurring in infection caused by several viruses. This narrative review explores the ADE phenomenon, its occurrence in viral infections and evaluates its role in infection by SARS-CoV-2 virus, which causes coronavirus disease 2019 (COVID-19). As of yet, there is no clear evidence of ADE in SARS-CoV-2, though this area is still subject to further study.
Collapse
Affiliation(s)
- Mohammed A. H. Farouq
- Department of Chemical and Process Engineering, University of Strathclyde, 75 Montrose Street, Glasgow G1 1XJ, UK; (P.A.M.); (M.M.A.Q.)
| | - Reinaldo Acevedo
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK;
| | - Valerie A. Ferro
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK;
| | - Paul A. Mulheran
- Department of Chemical and Process Engineering, University of Strathclyde, 75 Montrose Street, Glasgow G1 1XJ, UK; (P.A.M.); (M.M.A.Q.)
| | - Mohammed M. Al Qaraghuli
- Department of Chemical and Process Engineering, University of Strathclyde, 75 Montrose Street, Glasgow G1 1XJ, UK; (P.A.M.); (M.M.A.Q.)
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK;
- EPSRC Future Manufacturing Research Hub for Continuous Manufacturing and Advanced Crystallisation (CMAC), University of Strathclyde, 99 George Street, Glasgow G1 1RD, UK
| |
Collapse
|
10
|
Mammalian animal models for dengue virus infection: a recent overview. Arch Virol 2021; 167:31-44. [PMID: 34761286 PMCID: PMC8579898 DOI: 10.1007/s00705-021-05298-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/26/2021] [Indexed: 02/07/2023]
Abstract
Dengue, a rapidly spreading mosquito-borne human viral disease caused by dengue virus (DENV), is a public health concern in tropical and subtropical areas due to its expanding geographical range. DENV can cause a wide spectrum of illnesses in humans, ranging from asymptomatic infection or mild dengue fever (DF) to life-threatening dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). Dengue is caused by four DENV serotypes; however, dengue pathogenesis is complex and poorly understood. Establishing a useful animal model that can exhibit dengue-fever-like signs similar to those in humans is essential to improve our understanding of the host response and pathogenesis of DENV. Although several animal models, including mouse models, non-human primate models, and a recently reported tree shrew model, have been investigated for DENV infection, animal models with clinical signs that are similar to those of DF in humans have not yet been established. Although animal models are essential for understanding the pathogenesis of DENV infection and for drug and vaccine development, each animal model has its own strengths and limitations. Therefore, in this review, we provide a recent overview of animal models for DENV infection and pathogenesis, focusing on studies of the antibody-dependent enhancement (ADE) effect in animal models.
Collapse
|
11
|
Pinto AK, Hassert M, Han X, Barker D, Carnelley T, Branche E, Steffen TL, Stone ET, Geerling E, Viramontes KM, Nykiforuk C, Toth D, Shresta S, Kodihalli S, Brien JD. The Ability of Zika virus Intravenous Immunoglobulin to Protect From or Enhance Zika Virus Disease. Front Immunol 2021; 12:717425. [PMID: 34552587 PMCID: PMC8450494 DOI: 10.3389/fimmu.2021.717425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 08/09/2021] [Indexed: 11/21/2022] Open
Abstract
The closely related flaviviruses, dengue and Zika, cause significant human disease throughout the world. While cross-reactive antibodies have been demonstrated to have the capacity to potentiate disease or mediate protection during flavivirus infection, the mechanisms responsible for this dichotomy are still poorly understood. To understand how the human polyclonal antibody response can protect against, and potentiate the disease in the context of dengue and Zika virus infection we used intravenous hyperimmunoglobulin (IVIG) preparations in a mouse model of the disease. Three IVIGs (ZIKV-IG, Control-Ig and Gamunex®) were evaluated for their ability to neutralize and/or enhance Zika, dengue 2 and 3 viruses in vitro. The balance between virus neutralization and enhancement provided by the in vitro neutralization data was used to predict the IVIG concentrations which could protect or enhance Zika, and dengue 2 disease in vivo. Using this approach, we were able to define the unique in vivo dynamics of complex polyclonal antibodies, allowing for both enhancement and protection from flavivirus infection. Our results provide a novel understanding of how polyclonal antibodies interact with viruses with implications for the use of polyclonal antibody therapeutics and the development and evaluation of the next generation flavivirus vaccines.
Collapse
Affiliation(s)
- Amelia K. Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - Mariah Hassert
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - Xiaobing Han
- Emergent BioSolutions Canada Inc, Winnipeg, Canada
| | | | | | - Emilie Branche
- La Jolla Institute for Immunology, Center for Infectious Disease and Vaccine Research, La Jolla, CA, United States
| | - Tara L. Steffen
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - E. Taylor Stone
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - Karla M. Viramontes
- La Jolla Institute for Immunology, Center for Infectious Disease and Vaccine Research, La Jolla, CA, United States
| | | | - Derek Toth
- Emergent BioSolutions Canada Inc, Winnipeg, Canada
| | - Sujan Shresta
- La Jolla Institute for Immunology, Center for Infectious Disease and Vaccine Research, La Jolla, CA, United States
| | | | - James D. Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| |
Collapse
|
12
|
Prior Heterologous Flavivirus Exposure Results in Reduced Pathogenesis in a Mouse Model of Zika Virus Infection. J Virol 2021; 95:e0057321. [PMID: 34076486 PMCID: PMC8312874 DOI: 10.1128/jvi.00573-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The 2015/2016 Zika virus epidemic in South and Central America left the scientific community urgently trying to understand the factors that contribute to Zika virus pathogenesis. Because multiple other flaviviruses are endemic in areas where Zika virus emerged, it is hypothesized that a key to understanding Zika virus disease severity is to study Zika virus infection in the context of prior flavivirus exposure. Human and animal studies have highlighted the idea that having been previously exposed to a different flavivirus may modulate the immune response to Zika virus. However, it is still unclear how prior flavivirus exposure impacts Zika viral burden and disease. In this murine study, we longitudinally examine multiple factors involved in Zika disease, linking viral burden with increased neurological disease severity, weight loss, and inflammation. We show that prior heterologous flavivirus exposure with dengue virus type 2 or 3 or the vaccine strain of yellow fever provides protection from mortality in a lethal Zika virus challenge. However, reduction in viral burden and Zika disease varies depending on the infecting primary flavivirus; with primary Zika virus infection being most protective from Zika virus challenge, followed by dengue virus 2, with yellow fever and dengue virus 3 protecting against mortality but showing more severe disease. This study demonstrates the variation in protective effects of prior flavivirus exposure on Zika virus pathogenesis and identifies distinct relationships between primary flavivirus infection and the potential for Zika virus disease. IMPORTANCE The emergence and reemergence of various vector-borne diseases in recent years highlights the need to understand the mechanisms of protection for each pathogen. In this study, we investigated the impact of prior exposure to Zika virus, dengue virus serotypes 2 or 3, or the vaccine strain of yellow fever on pathogenesis and disease outcomes in a mouse model of Zika virus infection. We found that prior exposure to a heterologous flavivirus was protective from mortality, and to varying degrees, prior flavivirus exposure was protective against neurological disease, weight loss, and severe viral burden during a lethal Zika challenge. Using a longitudinal and cross-sectional study design, we were able to link multiple disease parameters, including viral burden, with neurological disease severity, weight loss, and inflammatory response in the context of flavivirus infection. This study demonstrates a measurable but varied impact of prior flavivirus exposure in modulating flavivirus pathophysiology. Given the cyclic nature of most flavivirus outbreaks, this work will contribute to the forecasting of disease severity for future outbreaks.
Collapse
|
13
|
Melo-Silva CR, Alves-Peixoto P, Heath N, Tang L, Montoya B, Knudson CJ, Stotesbury C, Ferez M, Wong E, Sigal LJ. Resistance to lethal ectromelia virus infection requires Type I interferon receptor in natural killer cells and monocytes but not in adaptive immune or parenchymal cells. PLoS Pathog 2021; 17:e1009593. [PMID: 34015056 PMCID: PMC8172060 DOI: 10.1371/journal.ppat.1009593] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 06/02/2021] [Accepted: 04/28/2021] [Indexed: 11/18/2022] Open
Abstract
Type I interferons (IFN-I) are antiviral cytokines that signal through the ubiquitous IFN-I receptor (IFNAR). Following footpad infection with ectromelia virus (ECTV), a mouse-specific pathogen, C57BL/6 (B6) mice survive without disease, while B6 mice broadly deficient in IFNAR succumb rapidly. We now show that for survival to ECTV, only hematopoietic cells require IFNAR expression. Survival to ECTV specifically requires IFNAR in both natural killer (NK) cells and monocytes. However, intrinsic IFNAR signaling is not essential for adaptive immune cell responses or to directly protect non-hematopoietic cells such as hepatocytes, which are principal ECTV targets. Mechanistically, IFNAR-deficient NK cells have reduced cytolytic function, while lack of IFNAR in monocytes dampens IFN-I production and hastens virus dissemination. Thus, during a pathogenic viral infection, IFN-I coordinates innate immunity by stimulating monocytes in a positive feedback loop and by inducing NK cell cytolytic function.
Collapse
Affiliation(s)
- Carolina R. Melo-Silva
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Pedro Alves-Peixoto
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Natasha Heath
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Lingjuan Tang
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Brian Montoya
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Cory J. Knudson
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Colby Stotesbury
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Maria Ferez
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Eric Wong
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Luis J. Sigal
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
14
|
Diagnostic differentiation of Zika and dengue virus exposure by analyzing T cell receptor sequences from peripheral blood of infected HLA-A2 transgenic mice. PLoS Negl Trop Dis 2020; 14:e0008896. [PMID: 33270635 PMCID: PMC7738164 DOI: 10.1371/journal.pntd.0008896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 12/15/2020] [Accepted: 10/15/2020] [Indexed: 11/19/2022] Open
Abstract
Zika virus (ZIKV) is a significant global health threat due to its potential for rapid emergence and association with severe congenital malformations during infection in pregnancy. Despite the urgent need, accurate diagnosis of ZIKV infection is still a major hurdle that must be overcome. Contributing to the inaccuracy of most serologically-based diagnostic assays for ZIKV, is the substantial geographic and antigenic overlap with other flaviviruses, including the four serotypes of dengue virus (DENV). Within this study, we have utilized a novel T cell receptor (TCR) sequencing platform to distinguish between ZIKV and DENV infections. Using high-throughput TCR sequencing of lymphocytes isolated from DENV and ZIKV infected mice, we were able to develop an algorithm which could identify virus-associated TCR sequences uniquely associated with either a prior ZIKV or DENV infection in mice. Using this algorithm, we were then able to separate mice that had been exposed to ZIKV or DENV infection with 97% accuracy. Overall this study serves as a proof-of-principle that T cell receptor sequencing can be used as a diagnostic tool capable of distinguishing between closely related viruses. Our results demonstrate the potential for this innovative platform to be used to accurately diagnose Zika virus infection and potentially the next emerging pathogen(s). Diagnostic differentiation between dengue virus and Zika virus infections is a challenge due to serological cross-reactivity. In this study, we used a novel T cell receptor sequencing platform to identify T cell receptor sequences significantly associated with either dengue or Zika virus infection in HLA-A2 transgenic mice. These libraries were used to computationally train diagnostic classifiers which were capable of distinguishing between dengue and Zika virus in independent cohorts of infected mice.
Collapse
|
15
|
Hassert M, Geerling E, Stone ET, Steffen TL, Feldman MS, Dickson AL, Class J, Richner JM, Brien JD, Pinto AK. mRNA induced expression of human angiotensin-converting enzyme 2 in mice for the study of the adaptive immune response to severe acute respiratory syndrome coronavirus 2. PLoS Pathog 2020; 16:e1009163. [PMID: 33326500 PMCID: PMC7773324 DOI: 10.1371/journal.ppat.1009163] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/30/2020] [Accepted: 11/23/2020] [Indexed: 12/20/2022] Open
Abstract
The novel human coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a pandemic. Critical to the rapid evaluation of vaccines and antivirals against SARS-CoV-2 is the development of tractable animal models to understand the adaptive immune response to the virus. To this end, the use of common laboratory strains of mice is hindered by significant divergence of the angiotensin-converting enzyme 2 (ACE2), which is the receptor required for entry of SARS-CoV-2. In the current study, we designed and utilized an mRNA-based transfection system to induce expression of the hACE2 receptor in order to confer entry of SARS-CoV-2 in otherwise non-permissive cells. By employing this expression system in an in vivo setting, we were able to interrogate the adaptive immune response to SARS-CoV-2 in type 1 interferon receptor deficient mice. In doing so, we showed that the T cell response to SARS-CoV-2 is enhanced when hACE2 is expressed during infection. Moreover, we demonstrated that these responses are preserved in memory and are boosted upon secondary infection. Importantly, using this system, we functionally identified the CD4+ and CD8+ structural peptide epitopes targeted during SARS-CoV-2 infection in H2b restricted mice and confirmed their existence in an established model of SARS-CoV-2 pathogenesis. We demonstrated that, identical to what has been seen in humans, the antigen-specific CD8+ T cells in mice primarily target peptides of the spike and membrane proteins, while the antigen-specific CD4+ T cells target peptides of the nucleocapsid, membrane, and spike proteins. As the focus of the immune response in mice is highly similar to that of the humans, the identification of functional murine SARS-CoV-2-specific T cell epitopes provided in this study will be critical for evaluation of vaccine efficacy in murine models of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Mariah Hassert
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, Missouri, United States of America
| | - Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, Missouri, United States of America
| | - E. Taylor Stone
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, Missouri, United States of America
| | - Tara L. Steffen
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, Missouri, United States of America
| | - Madi S. Feldman
- Department of Biomedical Engineering, Saint Louis University, St. Louis, Missouri, United States of America
| | - Alexandria L. Dickson
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, Missouri, United States of America
| | - Jacob Class
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Justin M. Richner
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - James D. Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, Missouri, United States of America
| | - Amelia K. Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, Missouri, United States of America
| |
Collapse
|
16
|
A Zika Vaccine Generated Using the Chimeric Insect-Specific Binjari Virus Platform Protects against Fetal Brain Infection in Pregnant Mice. Vaccines (Basel) 2020; 8:vaccines8030496. [PMID: 32887302 PMCID: PMC7564101 DOI: 10.3390/vaccines8030496] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 08/31/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023] Open
Abstract
Zika virus (ZIKV) is the etiological agent of congenital Zika syndrome (CZS), a spectrum of birth defects that can lead to life-long disabilities. A range of vaccines are in development with the target population including pregnant women and women of child-bearing age. Using a recently described chimeric flavivirus vaccine technology based on the novel insect-specific Binjari virus (BinJV), we generated a ZIKV vaccine (BinJ/ZIKA-prME) and illustrate herein its ability to protect against fetal brain infection. Female IFNAR−/− mice were vaccinated once with unadjuvanted BinJ/ZIKA-prME, were mated, and at embryonic day 12.5 were challenged with ZIKVPRVABC59. No infectious ZIKV was detected in maternal blood, placenta, or fetal heads in BinJ/ZIKA-prME-vaccinated mice. A similar result was obtained when the more sensitive qRT PCR methodology was used to measure the viral RNA. BinJ/ZIKA-prME vaccination also did not result in antibody-dependent enhancement of dengue virus infection or disease. BinJ/ZIKA-prME thus emerges as a potential vaccine candidate for the prevention of CSZ.
Collapse
|
17
|
Bournazos S, Gupta A, Ravetch JV. The role of IgG Fc receptors in antibody-dependent enhancement. Nat Rev Immunol 2020; 20:633-643. [PMID: 32782358 PMCID: PMC7418887 DOI: 10.1038/s41577-020-00410-0] [Citation(s) in RCA: 345] [Impact Index Per Article: 86.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2020] [Indexed: 02/07/2023]
Abstract
Antibody-dependent enhancement (ADE) is a mechanism by which the pathogenesis of certain viral infections is enhanced in the presence of sub-neutralizing or cross-reactive non-neutralizing antiviral antibodies. In vitro modelling of ADE has attributed enhanced pathogenesis to Fcγ receptor (FcγR)-mediated viral entry, rather than canonical viral receptor-mediated entry. However, the putative FcγR-dependent mechanisms of ADE overlap with the role of these receptors in mediating antiviral protection in various viral infections, necessitating a detailed understanding of how this diverse family of receptors functions in protection and pathogenesis. Here, we discuss the diversity of immune responses mediated upon FcγR engagement and review the available experimental evidence supporting the role of FcγRs in antiviral protection and pathogenesis through ADE. We explore FcγR engagement in the context of a range of different viral infections, including dengue virus and SARS-CoV, and consider ADE in the context of the ongoing SARS-CoV-2 pandemic. Antibody-dependent enhancement (ADE) has been described as a mechanism that contributes to the pathogenesis of dengue virus infection. Limited evidence also suggests that it can also occur in other viral infections. Here, the authors explore the history of the ADE phenomenon, discuss the diversity of Fc effector functions and consider its potential relevance in the context of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Stylianos Bournazos
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY, USA
| | - Aaron Gupta
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY, USA
| | - Jeffrey V Ravetch
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
18
|
Hassert M, Geerling E, Stone ET, Steffen TL, Dickson A, Feldman MS, Class J, Richner JM, Brien JD, Pinto AK. mRNA induced expression of human angiotensin-converting enzyme 2 in mice for the study of the adaptive immune response to severe acute respiratory syndrome coronavirus 2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32793909 DOI: 10.1101/2020.08.07.241877] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The novel human coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a pandemic resulting in nearly 20 million infections across the globe, as of August 2020. Critical to the rapid evaluation of vaccines and antivirals is the development of tractable animal models of infection. The use of common laboratory strains of mice to this end is hindered by significant divergence of the angiotensin-converting enzyme 2 (ACE2), which is the receptor required for entry of SARS-CoV-2. In the current study, we designed and utilized an mRNA-based transfection system to induce expression of the hACE2 receptor in order to confer entry of SARS-CoV-2 in otherwise non-permissive cells. By employing this expression system in an in vivo setting, we were able to interrogate the adaptive immune response to SARS-CoV-2 in type 1 interferon receptor deficient mice. In doing so, we showed that the T cell response to SARS-CoV-2 is enhanced when hACE2 is expressed during infection. Moreover, we demonstrated that these responses are preserved in memory and are boosted upon secondary infection. Interestingly, we did not observe an enhancement of SARS-CoV-2 specific antibody responses with hACE2 induction. Importantly, using this system, we functionally identified the CD4+ and CD8+ peptide epitopes targeted during SARS-CoV-2 infection in H2b restricted mice. Antigen-specific CD8+ T cells in mice of this MHC haplotype primarily target peptides of the spike and membrane proteins, while the antigen-specific CD4+ T cells target peptides of the nucleocapsid, membrane, and spike proteins. The functional identification of these T cell epitopes will be critical for evaluation of vaccine efficacy in murine models of SARS-CoV-2. The use of this tractable expression system has the potential to be used in other instances of emerging infections in which the rapid development of an animal model is hindered by a lack of host susceptibility factors.
Collapse
|
19
|
Chen RE, Diamond MS. Dengue mouse models for evaluating pathogenesis and countermeasures. Curr Opin Virol 2020; 43:50-58. [PMID: 32950933 PMCID: PMC7774505 DOI: 10.1016/j.coviro.2020.09.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/01/2020] [Indexed: 12/14/2022]
Abstract
Dengue virus (DENV) causes the most prevalent arbovirus illness worldwide and is responsible for many debilitating epidemics. The four circulating DENV serotypes infect humans and can cause asymptomatic, mild, moderate, or severe Dengue. Because of the global morbidity and mortality due to Dengue, deployment of a safe and effective tetravalent vaccine has been a high priority, and to date, a partially realized goal. The study of pathogenesis and development of DENV therapeutics and vaccines has been limited by few animal models that recapitulate key features of human disease. Over the past two decades, mouse models of DENV infection have evolved with increasing success. Here, we review the utilization and limitations of mice for studying DENV pathogenesis and evaluating countermeasures.
Collapse
Affiliation(s)
- Rita E Chen
- 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
| | - 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; The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
20
|
The small molecule AZD6244 inhibits dengue virus replication in vitro and protects against lethal challenge in a mouse model. Arch Virol 2020; 165:671-681. [PMID: 31942645 DOI: 10.1007/s00705-020-04524-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/06/2019] [Indexed: 12/12/2022]
Abstract
Dengue virus (DENV) is the most common mosquito-borne viral disease. The World Health Organization estimates that 400 million new cases of dengue fever occur every year. Approximately 500,000 individuals develop severe and life-threatening complications from dengue fever, such as dengue shock syndrome (DSS) and dengue hemorrhagic fever (DHF), which cause 22,000 deaths yearly. Currently, there are no specific licensed therapeutics to treat DENV illness. We have previously shown that the MEK/ERK inhibitor U0126 inhibits the replication of the flavivirus yellow fever virus. In this study, we demonstrate that the MEK/ERK inhibitor AZD6244 has potent antiviral efficacy in vitro against DENV-2, DENV-3, and Saint Louis encephalitis virus (SLEV). We also show that it is able to protect AG129 mice from a lethal challenge with DENV-2 (D2S20). The molecule is currently undergoing phase III clinical trials for the treatment of non-small-cell lung cancer. The effect of AZD6244 on the DENV life cycle was attributed to a blockade of morphogenesis. Treatment of AG129 mice twice daily with oral doses of AZD6244 (100 mg/kg/day) prevented the animals from contracting dengue hemorrhagic fever (DHF)-like lethal disease upon intravenous infection with 1 × 105 PFU of D2S20. The effectiveness of AZD6244 was observed even when the treatment of infected animals was initiated 1-2 days postinfection. This was also followed by a reduction in viral copy number in both the serum and the spleen. There was also an increase in IL-1β and TNF-α levels in mice that were infected with D2S20 and treated with AZD6244 in comparison to infected mice that were treated with the vehicle only. These data demonstrate the potential of AZD6244 as a new therapeutic agent to treat DENV infection and possibly other flavivirus diseases.
Collapse
|
21
|
Thackray LB, Handley SA, Gorman MJ, Poddar S, Bagadia P, Briseño CG, Theisen DJ, Tan Q, Hykes BL, Lin H, Lucas TM, Desai C, Gordon JI, Murphy KM, Virgin HW, Diamond MS. Oral Antibiotic Treatment of Mice Exacerbates the Disease Severity of Multiple Flavivirus Infections. Cell Rep 2019; 22:3440-3453.e6. [PMID: 29590614 PMCID: PMC5908250 DOI: 10.1016/j.celrep.2018.03.001] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 02/15/2018] [Accepted: 02/28/2018] [Indexed: 12/22/2022] Open
Abstract
Although the outcome of flavivirus infection can vary from asymptomatic to lethal, environmental factors modulating disease severity are poorly defined. Here, we observed increased susceptibility of mice to severe West Nile (WNV), Dengue, and Zika virus infections after treatment with oral antibiotics (Abx) that depleted the gut microbiota. Abx treatment impaired the development of optimal T cell responses, with decreased levels of WNV-specific CD8+ T cells associated with increased infection and immunopathology. Abx treatments that resulted in enhanced WNV susceptibility generated changes in the overall structure of the gut bacterial community and in the abundance of specific bacterial taxa. As little as 3 days of treatment with ampicillin was sufficient to alter host immunity and WNV outcome. Our results identify oral Abx therapy as a potential environmental determinant of systemic viral disease, and they raise the possibility that perturbation of the gut microbiota may have deleterious consequences for subsequent flavivirus infections.
Collapse
Affiliation(s)
- Larissa B Thackray
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Scott A Handley
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Matthew J Gorman
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Subhajit Poddar
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Prachi Bagadia
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Carlos G Briseño
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Derek J Theisen
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Qing Tan
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Barry L Hykes
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Hueylie Lin
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Tiffany M Lucas
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Chandni Desai
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jeffrey I Gordon
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Howard Hughes Medical Institute, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Herbert W Virgin
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
22
|
Fowler AM, Tang WW, Young MP, Mamidi A, Viramontes KM, McCauley MD, Carlin AF, Schooley RT, Swanstrom J, Baric RS, Govero J, Diamond MS, Shresta S. Maternally Acquired Zika Antibodies Enhance Dengue Disease Severity in Mice. Cell Host Microbe 2019; 24:743-750.e5. [PMID: 30439343 DOI: 10.1016/j.chom.2018.09.015] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 06/21/2018] [Accepted: 09/20/2018] [Indexed: 01/08/2023]
Abstract
Antibody (Ab)-dependent enhancement can exacerbate dengue virus (DENV) infection due to cross-reactive Abs from an initial DENV infection, facilitating replication of a second DENV. Zika virus (ZIKV) emerged in DENV-endemic areas, raising questions about whether existing immunity could affect these related flaviviruses. We show that mice born with circulating maternal Abs against ZIKV develop severe disease upon DENV infection. Compared with pups of naive mothers, those born to ZIKV-immune mice lacking type I interferon receptor in myeloid cells (LysMCre+Ifnar1fl/fl) exhibit heightened disease and viremia upon DENV infection. Passive transfer of IgG isolated from mice born to ZIKV-immune mothers resulted in increased viremia in naive recipient mice. Treatment with Abs blocking inflammatory cytokine tumor necrosis factor linked to DENV disease or Abs blocking DENV entry improved survival of DENV-infected mice born to ZIKV-immune mothers. Thus, the maternal Ab response to ZIKV infection or vaccination might predispose to severe dengue disease in infants.
Collapse
Affiliation(s)
- Angela M Fowler
- Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - William W Tang
- Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Matthew P Young
- Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Anila Mamidi
- Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Karla M Viramontes
- Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Melanie D McCauley
- Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Aaron F Carlin
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Robert T Schooley
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jesica Swanstrom
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jennifer Govero
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sujan Shresta
- Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA; Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
23
|
Hassert M, Brien JD, Pinto AK. Mouse Models of Heterologous Flavivirus Immunity: A Role for Cross-Reactive T Cells. Front Immunol 2019; 10:1045. [PMID: 31143185 PMCID: PMC6520664 DOI: 10.3389/fimmu.2019.01045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 04/24/2019] [Indexed: 12/20/2022] Open
Abstract
Most of the world is at risk of being infected with a flavivirus such as dengue virus, West Nile virus, yellow fever virus, Japanese encephalitis virus, tick-borne encephalitis virus, and Zika virus, significantly impacting millions of lives. Importantly, many of these genetically similar viruses co-circulate within the same geographic regions, making it likely for individuals living in areas of high flavivirus endemicity to be infected with multiple flaviviruses during their lifetime. Following a flavivirus infection, a robust virus-specific T cell response is generated and the memory recall of this response has been demonstrated to provide long-lasting immunity, protecting against reinfection with the same pathogen. However, multiple studies have shown that this flavivirus specific T cell response can be cross-reactive and active during heterologous flavivirus infection, leading to the question: How does immunity to one flavivirus shape immunity to the next, and how does this impact disease? It has been proposed that in some cases unfavorable disease outcomes may be caused by lower avidity cross-reactive memory T cells generated during a primary flavivirus infection that preferentially expand during a secondary heterologous infection and function sub optimally against the new pathogen. While in other cases, these cross-reactive cells still have the potential to facilitate cross-protection. In this review, we focus on cross-reactive T cell responses to flaviviruses and the concepts and consequences of T cell cross-reactivity, with particular emphasis linking data generated using murine models to our new understanding of disease outcomes following heterologous flavivirus infection.
Collapse
Affiliation(s)
- Mariah Hassert
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, United States
| | - James D Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, United States
| | - Amelia K Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, United States
| |
Collapse
|
24
|
The Temporal Role of Cytokines in Flavivirus Protection and Pathogenesis. CURRENT CLINICAL MICROBIOLOGY REPORTS 2018. [DOI: 10.1007/s40588-018-0106-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
25
|
Abstract
An infection by Zika virus (ZIKV), a mosquito-borne flavivirus, broke out in South American regions in 2015, and recently showed a tendency of spreading to North America and even worldwide. ZIKV was first detected in 1947 and only 14 human infection cases were reported until 2007. This virus was previously observed to cause only mild flu-like symptoms. However, recent ZIKV infections might be responsible for the increasing cases of neurological disorders such as Guillain-Barré syndrome and congenital defects, including newborn microcephaly. Therefore, researchers have established several animal models to study ZIKV transmission and pathogenesis, and test therapeutic candidates. This review mainly summarizes the reported animal models of ZIKV infection, including mice and non-human primates.
Collapse
|
26
|
Abstract
Flaviviruses such as dengue (DENV), yellow fever (YFV), West Nile (WNV), and Zika (ZIKV) are human pathogens of global significance. In particular, DENV causes the most prevalent mosquito-borne viral diseases in humans, and ZIKV emerged from obscurity into the spotlight in 2016 as the etiologic agent of congenital Zika syndrome. Owing to the recent emergence of ZIKV as a global pandemic threat, the roles of the immune system during ZIKV infections are as yet unclear. In contrast, decades of DENV research implicate a dual role for the immune system in protection against and pathogenesis of DENV infection. As DENV and ZIKV are closely related, knowledge based on DENV studies has been used to prioritize investigation of ZIKV immunity and pathogenesis, and to accelerate ZIKV diagnostic, therapeutic, and vaccine design. This review discusses the following topics related to innate and adaptive immune responses to DENV and ZIKV: the interferon system as the key mechanism of host defense and viral target for immune evasion, antibody-mediated protection versus antibody-dependent enhancement, and T cell-mediated protection versus original T cell antigenic sin. Understanding the mechanisms that regulate the balance between immune-mediated protection and pathogenesis during DENV and ZIKV infections is critical toward development of safe and effective DENV and ZIKV therapeutics and vaccines.
Collapse
Affiliation(s)
- Annie Elong Ngono
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA;
| | - Sujan Shresta
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA;
| |
Collapse
|
27
|
Animal Models for Dengue and Zika Vaccine Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1062:215-239. [PMID: 29845536 DOI: 10.1007/978-981-10-8727-1_16] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The current status of animal models in the study of dengue and Zika are covered in this review. Mouse models deficient in IFN signaling are used to overcome the natural resistance of mice to non-encephalitic flaviviruses. Conditional IFNAR mice and non-human primates (NHP) are useful immuno-competent models. Sterile immunity after dengue vaccination is not observed in NHPs. Placental and fetal development in NHPs is similar to humans, facilitating studies on infection-mediated fetal impairment.
Collapse
|
28
|
Abstract
A recent unprecedented outbreak of Zika virus (ZIKV) in the Americas has been associated with microcephaly and other congenital malformations in infants as well as Guillain-Barre syndrome in adults. The development of a safe and effective ZIKV vaccine is therefore an urgent global health priority. Promising data from preclinical vaccine studies in mice and monkeys suggest that an effective vaccine will likely be possible, but important scientific challenges remain. Here we review the current state of ZIKV vaccine development. We discuss different vaccination strategies and we highlight challenges facing clinical evaluation of ZIKV vaccine candidates.
Collapse
Affiliation(s)
- Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.
| | - Stephen J Thomas
- Upstate Medical University, State University of New York, Syracuse, NY 13210, USA
| | - Nelson L Michael
- Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| |
Collapse
|
29
|
Tick-borne encephalitis virus neutralization by high dose intravenous immunoglobulin. Ticks Tick Borne Dis 2017; 8:253-258. [DOI: 10.1016/j.ttbdis.2016.11.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 11/08/2016] [Accepted: 11/14/2016] [Indexed: 11/22/2022]
|
30
|
The Many Faces of the Flavivirus NS5 Protein in Antagonism of Type I Interferon Signaling. J Virol 2017; 91:JVI.01970-16. [PMID: 27881649 DOI: 10.1128/jvi.01970-16] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The vector-borne flaviviruses cause severe disease in humans on every inhabited continent on earth. Their transmission by arthropods, particularly mosquitoes, facilitates large emergence events such as witnessed with Zika virus (ZIKV) or West Nile virus in the Americas. Every vector-borne flavivirus examined thus far that causes disease in humans, from dengue virus to ZIKV, antagonizes the host type I interferon (IFN-I) response by preventing JAK-STAT signaling, suggesting that suppression of this pathway is an important determinant of infection. The most direct and potent viral inhibitor of this pathway is the nonstructural protein NS5. However, the mechanisms utilized by NS5 from different flaviviruses are often quite different, sometimes despite close evolutionary relationships between viruses. The varied mechanisms of NS5 as an IFN-I antagonist are also surprising given that the evolution of NS5 is restrained by the requirement to maintain function of two enzymatic activities critical for virus replication, the methyltransferase and RNA-dependent RNA polymerase. This review discusses the different strategies used by flavivirus NS5 to evade the antiviral effects of IFN-I and how this information can be used to better model disease and develop antiviral countermeasures.
Collapse
|
31
|
Lai JH, Lin YL, Hsieh SL. Pharmacological intervention for dengue virus infection. Biochem Pharmacol 2017; 129:14-25. [PMID: 28104437 DOI: 10.1016/j.bcp.2017.01.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 01/12/2017] [Indexed: 12/11/2022]
Abstract
Dengue virus (DENV) infection has a considerable health impact in tropical and subtropical countries worldwide. Escalation of infection rates greatly increases morbidity and mortality, most commonly from deaths due to dengue hemorrhagic fever and dengue shock syndrome. Although the development of an effective, long-lasting vaccine has been a major aim for control and prevention of DENV infection, the currently licensed vaccine has limitations and is less than satisfactory. Thus, there remains an important need to identify effective and tolerable medications for treatment of DENV-infected patients both in the early phase, to prevent progression to fatal outcomes, and to minimize deaths after patients develop severe complications. This review will address several specific points, including (1) approaches to identify anti-DENV medications, (2) recent advances in the development of potential compounds targeting DENV infection, (3) experience with clinical trials of regimens for DENV infection, (4) some available medications of potential for clinical trials against DENV infection, (5) reasons for unsuccessful outcomes and challenges of anti-DENV treatments, and (6) directions for developing or selecting better anti-DENV strategies. This review provides useful guidance for clinicians selecting drugs for DENV-infected patients with severe manifestations or potential fatal disease progression, and for basic researchers seeking to develop effective anti-DENV regimens.
Collapse
Affiliation(s)
- Jenn-Haung Lai
- Division of Rheumatology, Allergy, and Immunology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Tao-Yuan, Taiwan, ROC; Graduate Institute of Medical Science, National Defense Medical Center, Taipei, Taiwan, ROC.
| | - Yi-Ling Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
| | - Shie-Liang Hsieh
- Institute of Microbiology and Immunology, National Yang-Ming University, Taiwan, ROC; Institute of Clinical Medicine, National Yang-Ming University, Taiwan, ROC; Genomics Research Center, Academia Sinica, Taipei, Taiwan, ROC; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| |
Collapse
|
32
|
Elong Ngono A, Vizcarra EA, Tang WW, Sheets N, Joo Y, Kim K, Gorman MJ, Diamond MS, Shresta S. Mapping and Role of the CD8 + T Cell Response During Primary Zika Virus Infection in Mice. Cell Host Microbe 2017; 21:35-46. [PMID: 28081442 PMCID: PMC5234855 DOI: 10.1016/j.chom.2016.12.010] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 11/15/2016] [Accepted: 12/15/2016] [Indexed: 11/26/2022]
Abstract
CD8+ T cells may play a dual role in protection against and pathogenesis of flaviviruses, including Zika virus (ZIKV). We evaluated the CD8+ T cell response in ZIKV-infected LysMCre+IFNARfl/fl C57BL/6 (H-2b) mice lacking the type I interferon receptor in a subset of myeloid cells. In total, 26 and 15 CD8+ T cell-reactive peptides for ZIKV African (MR766) and Asian (FSS13025) lineage strains, respectively, were identified and validated. CD8+ T cells from infected mice were polyfunctional and mediated cytotoxicity. Adoptive transfer of ZIKV-immune CD8+ T cells reduced viral burdens, whereas their depletion led to higher tissue burdens, and CD8-/- mice displayed higher mortality with ZIKV infection. Collectively, these results demonstrate that CD8+ T cells protect against ZIKV infection. Further, this study provides a T cell competent mouse model for investigating ZIKV-specific T cell responses.
Collapse
Affiliation(s)
- Annie Elong Ngono
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Edward A Vizcarra
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - William W Tang
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Nicholas Sheets
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Yunichel Joo
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Kenneth Kim
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Matthew J Gorman
- Department of Medicine, Molecular Microbiology, Pathology, and Immunology, Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael S Diamond
- Department of Medicine, Molecular Microbiology, Pathology, and Immunology, Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sujan Shresta
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.
| |
Collapse
|
33
|
Liu Y, Olagnier D, Lin R. Host and Viral Modulation of RIG-I-Mediated Antiviral Immunity. Front Immunol 2017; 7:662. [PMID: 28096803 PMCID: PMC5206486 DOI: 10.3389/fimmu.2016.00662] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/16/2016] [Indexed: 12/21/2022] Open
Abstract
Innate immunity is the first line of defense against invading pathogens. Rapid and efficient detection of pathogen-associated molecular patterns via pattern-recognition receptors is essential for the host to mount defensive and protective responses. Retinoic acid-inducible gene-I (RIG-I) is critical in triggering antiviral and inflammatory responses for the control of viral replication in response to cytoplasmic virus-specific RNA structures. Upon viral RNA recognition, RIG-I recruits the mitochondrial adaptor protein mitochondrial antiviral signaling protein, which leads to a signaling cascade that coordinates the induction of type I interferons (IFNs), as well as a large variety of antiviral interferon-stimulated genes. The RIG-I activation is tightly regulated via various posttranslational modifications for the prevention of aberrant innate immune signaling. By contrast, viruses have evolved mechanisms of evasion, such as sequestrating viral structures from RIG-I detections and targeting receptor or signaling molecules for degradation. These virus–host interactions have broadened our understanding of viral pathogenesis and provided insights into the function of the RIG-I pathway. In this review, we summarize the recent advances regarding RIG-I pathogen recognition and signaling transduction, cell-intrinsic control of RIG-I activation, and the viral antagonism of RIG-I signaling.
Collapse
Affiliation(s)
- Yiliu Liu
- Jewish General Hospital, Lady Davis Institute, McGill University, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - David Olagnier
- Jewish General Hospital, Lady Davis Institute, McGill University, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Rongtuan Lin
- Jewish General Hospital, Lady Davis Institute, McGill University, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada; Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| |
Collapse
|
34
|
Towards antiviral therapies for treating dengue virus infections. Curr Opin Pharmacol 2016; 30:1-7. [DOI: 10.1016/j.coph.2016.06.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 06/08/2016] [Indexed: 01/27/2023]
|
35
|
Makhluf H, Kim K, Shresta S. Novel strategies for discovering inhibitors of Dengue and Zika fever. Expert Opin Drug Discov 2016; 11:921-3. [DOI: 10.1080/17460441.2016.1212013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
36
|
Miner JJ, Cao B, Govero J, Smith AM, Fernandez E, Cabrera OH, Garber C, Noll M, Klein RS, Noguchi KK, Mysorekar IU, Diamond MS. Zika Virus Infection during Pregnancy in Mice Causes Placental Damage and Fetal Demise. Cell 2016; 165:1081-1091. [PMID: 27180225 PMCID: PMC4874881 DOI: 10.1016/j.cell.2016.05.008] [Citation(s) in RCA: 647] [Impact Index Per Article: 80.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 05/02/2016] [Accepted: 05/02/2016] [Indexed: 12/25/2022]
Abstract
Zika virus (ZIKV) infection in pregnant women causes intrauterine growth restriction, spontaneous abortion, and microcephaly. Here, we describe two mouse models of placental and fetal disease associated with in utero transmission of ZIKV. Female mice lacking type I interferon signaling (Ifnar1(-/-)) crossed to wild-type (WT) males produced heterozygous fetuses resembling the immune status of human fetuses. Maternal inoculation at embryonic day 6.5 (E6.5) or E7.5 resulted in fetal demise that was associated with ZIKV infection of the placenta and fetal brain. We identified ZIKV within trophoblasts of the maternal and fetal placenta, consistent with a trans-placental infection route. Antibody blockade of Ifnar1 signaling in WT pregnant mice enhanced ZIKV trans-placental infection although it did not result in fetal death. These models will facilitate the study of ZIKV pathogenesis, in utero transmission, and testing of therapies and vaccines to prevent congenital malformations.
Collapse
Affiliation(s)
- Jonathan J Miner
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Bin Cao
- Department of Obstetrics and Gynecology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jennifer Govero
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Amber M Smith
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Estefania Fernandez
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Omar H Cabrera
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Charise Garber
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Michelle Noll
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Robyn S Klein
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Anatomy and Neurobiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Kevin K Noguchi
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Indira U Mysorekar
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Obstetrics and Gynecology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; The Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
37
|
Lazear HM, Govero J, Smith AM, Platt DJ, Fernandez E, Miner JJ, Diamond MS. A Mouse Model of Zika Virus Pathogenesis. Cell Host Microbe 2016; 19:720-30. [PMID: 27066744 PMCID: PMC4866885 DOI: 10.1016/j.chom.2016.03.010] [Citation(s) in RCA: 719] [Impact Index Per Article: 89.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 03/28/2016] [Accepted: 03/28/2016] [Indexed: 12/11/2022]
Abstract
The ongoing Zika virus (ZIKV) epidemic and unexpected clinical outcomes, including Guillain-Barré syndrome and birth defects, has brought an urgent need for animal models. We evaluated infection and pathogenesis with contemporary and historical ZIKV strains in immunocompetent mice and mice lacking components of the antiviral response. Four- to six-week-old Irf3(-/-)Irf5(-/-)Irf7(-/-) triple knockout mice, which produce little interferon α/β, and mice lacking the interferon receptor (Ifnar1(-/-)) developed neurological disease and succumbed to ZIKV infection, whereas single Irf3(-/-), Irf5(-/-), and Mavs(-/-) knockout mice exhibited no overt illness. Ifnar1(-/-) mice sustained high viral loads in the brain and spinal cord, consistent with evidence that ZIKV causes neurodevelopmental defects in human fetuses. The testes of Ifnar1(-/-) mice had the highest viral loads, which is relevant to sexual transmission of ZIKV. This model of ZIKV pathogenesis will be valuable for evaluating vaccines and therapeutics as well as understanding disease pathogenesis.
Collapse
Affiliation(s)
- Helen M Lazear
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jennifer Govero
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Amber M Smith
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Derek J Platt
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Estefania Fernandez
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jonathan J Miner
- Department of Medicine, 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 Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; The Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
38
|
Olagnier D, Amatore D, Castiello L, Ferrari M, Palermo E, Diamond MS, Palamara AT, Hiscott J. Dengue Virus Immunopathogenesis: Lessons Applicable to the Emergence of Zika Virus. J Mol Biol 2016; 428:3429-48. [PMID: 27130436 DOI: 10.1016/j.jmb.2016.04.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 04/14/2016] [Accepted: 04/16/2016] [Indexed: 01/07/2023]
Abstract
Dengue is the leading mosquito-transmitted viral infection in the world. There are more than 390 million new infections annually; while the majority of infected individuals are asymptomatic or develop a self-limited dengue fever, up to 1 million clinical cases develop severe manifestations, including dengue hemorrhagic fever and shock syndrome, resulting in ~25,000 deaths annually, mainly in children. Gaps in our understanding of the mechanisms that contribute to dengue infection and immunopathogenesis have hampered the development of vaccines and antiviral agents. Some of these limitations are highlighted by the explosive re-emergence of another arthropod-borne flavivirus-Zika virus-spread by the same vector, the Aedes aegypti mosquito, that also carries dengue, yellow fever and chikungunya viruses. This review will discuss the early virus-host interactions in dengue infection, with emphasis on the interrelationship between oxidative stress and innate immune pathways, and will provide insight as to how lessons learned from dengue research may expedite therapeutic strategies for Zika virus.
Collapse
Affiliation(s)
- David Olagnier
- Lady Davis Institute, Jewish General Hospital, McGill University Montreal, Canada
| | - Donatella Amatore
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | | | - Matteo Ferrari
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| | - Enrico Palermo
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University at St. Louis, St. Louis, MO, USA
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy; Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| | - John Hiscott
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy.
| |
Collapse
|
39
|
Martínez Gómez JM, Ong LC, Lam JH, Binte Aman SA, Libau EA, Lee PX, St. John AL, Alonso S. Maternal Antibody-Mediated Disease Enhancement in Type I Interferon-Deficient Mice Leads to Lethal Disease Associated with Liver Damage. PLoS Negl Trop Dis 2016; 10:e0004536. [PMID: 27007501 PMCID: PMC4805191 DOI: 10.1371/journal.pntd.0004536] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 02/22/2016] [Indexed: 11/18/2022] Open
Abstract
Epidemiological studies have reported that most of the severe dengue cases occur upon a secondary heterologous infection. Furthermore, babies born to dengue immune mothers are at greater risk of developing severe disease upon primary infection with a heterologous or homologous dengue virus (DENV) serotype when maternal antibodies reach sub-neutralizing concentrations. These observations have been explained by the antibody mediated disease enhancement (ADE) phenomenon whereby heterologous antibodies or sub-neutralizing homologous antibodies bind to but fail to neutralize DENV particles, allowing Fc-receptor mediated entry of the virus-antibody complexes into host cells. This eventually results in enhanced viral replication and heightened inflammatory responses. In an attempt to replicate this ADE phenomenon in a mouse model, we previously reported that upon DENV2 infection 5-week old type I and II interferon (IFN) receptors-deficient mice (AG129) born to DENV1-immune mothers displayed enhancement of disease severity characterized by increased virus titers and extensive vascular leakage which eventually led to the animals' death. However, as dengue occurs in immune competent individuals, we sought to reproduce this mouse model in a less immunocompromised background. Here, we report an ADE model that is mediated by maternal antibodies in type I IFN receptor-deficient A129 mice. We show that 5-week old A129 mice born to DENV1-immune mothers succumbed to a DENV2 infection within 4 days that was sub-lethal in mice born to naïve mothers. Clinical manifestations included extensive hepatocyte vacuolation, moderate vascular leakage, lymphopenia, and thrombocytopenia. Anti-TNFα therapy totally protected the mice and correlated with healthy hepatocytes. In contrast, blocking IL-6 did not impact the virus titers or disease outcome. This A129 mouse model of ADE may help dissecting the mechanisms involved in dengue pathogenesis and evaluate the efficacy of vaccine and therapeutic candidates.
Collapse
Affiliation(s)
- Julia María Martínez Gómez
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Li Ching Ong
- Infectious Disease programme, Singapore-MIT alliance for Research and Technology (SMART), National University of Singapore, Singapore
| | - Jian Hang Lam
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology programme, Life Sciences Institute, National University of Singapore, Singapore
| | | | - Eshele Anak Libau
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Pei Xuan Lee
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology programme, Life Sciences Institute, National University of Singapore, Singapore
| | | | - Sylvie Alonso
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology programme, Life Sciences Institute, National University of Singapore, Singapore
- Infectious Disease programme, Singapore-MIT alliance for Research and Technology (SMART), National University of Singapore, Singapore
- * E-mail:
| |
Collapse
|
40
|
Moi ML, Takasaki T, Kurane I. Human antibody response to dengue virus: implications for dengue vaccine design. Trop Med Health 2016; 44:1. [PMID: 27398060 PMCID: PMC4934144 DOI: 10.1186/s41182-016-0004-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 01/13/2016] [Indexed: 11/10/2022] Open
Abstract
Dengue, a global health threat, is a leading cause of morbidity and mortality in most tropical and subtropical countries. Dengue can range from asymptomatic, relatively mild dengue fever to severe and life-threatening dengue hemorrhagic fever. Disease severity and outcome is largely associated with the host immune response. Several candidate vaccines in clinical trials appear promising as effective measures for dengue disease control. Vaccine development has been hampered by safety and efficacy issues, driven by a lack of understanding of the host immune response. This review focuses on recent research findings on the dengue host immune response, particularly in humans, and the relevance of these findings to challenges in vaccine development.
Collapse
Affiliation(s)
- Meng Ling Moi
- />Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
- />National Institute of Infectious Diseases, Tokyo, Japan
| | | | - Ichiro Kurane
- />National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|