1
|
Melendez JA, Sun H, Bonner J, Chen Q. Characterization of a plant-derived monoclonal antibody targeting extracellular enveloped virions of Monkeypox virus. FRONTIERS IN PLANT SCIENCE 2024; 15:1481452. [PMID: 39554528 PMCID: PMC11563991 DOI: 10.3389/fpls.2024.1481452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/10/2024] [Indexed: 11/19/2024]
Abstract
In 2022, the global outbreak of monkeypox virus (MPXV) with increased human-to-human transmission triggered urgent public health interventions. Plant-derived monoclonal antibodies (mAbs) are being explored as potential therapeutic strategies due to their diverse mechanisms of antiviral activity. MPXV produces two key infectious particles: the mature virion (MV) and the extracellular enveloped virion (EV), both essential for infection and spread. Effective therapies must target both to halt replication and transmission. Our prior research demonstrated the development of a potent neutralizing mAb against MPXV MV. This study focuses on developing a plant-derived mAb targeting MPXV EV, which is critical for viral dissemination within the host and generally resistant to antibody neutralization. Our findings reveal that the mAb (H2) can be robustly produced in Nicotiana benthamiana plants via transient expression. The plant-made H2 mAb effectively targets MPXV EV by binding specifically to the A35 MPXV antigen. Importantly, H2 mAb shows notable neutralizing activity against the infectious MPXV EV particle. This investigation is the first to report the development of a plant-derived anti-EV mAb for MPXV prevention and treatment, as well as the first demonstration of anti-MPXV EV activity by an mAb across any production platform. It highlights the potential of plant-produced mAbs as therapeutics for emerging infectious diseases, including the MPXV outbreak.
Collapse
Affiliation(s)
- Jennifer A Melendez
- Biodesign Institute, Arizona State University, Tempe, AZ, United States
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Haiyan Sun
- Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - James Bonner
- Biodesign Institute, Arizona State University, Tempe, AZ, United States
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Qiang Chen
- Biodesign Institute, Arizona State University, Tempe, AZ, United States
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
2
|
Krittanai S, Rattanapisit K, Bulaon CJI, Pitaksajjakul P, Keadsanti S, Ramasoota P, Strasser R, Phoolcharoen W. Nicotiana benthamiana as a potential source for producing anti-dengue virus D54 neutralizing therapeutic antibody. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2024; 42:e00844. [PMID: 38881650 PMCID: PMC11179242 DOI: 10.1016/j.btre.2024.e00844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 05/07/2024] [Accepted: 05/26/2024] [Indexed: 06/18/2024]
Abstract
Dengue virus (DENV), transmitted by mosquitoes, is classified into four serotypes (DENV1-4) and typically causes mild, self-limiting symptoms upon initial infection. However, secondary infection can lead to severe symptoms due to antibody-dependent enhancement (ADE). To address this, anti-DENV antibodies are being developed with the goal of neutralizing infection without ADE activity. Previous attempts using a 54_hG1 antibody from CHO-K1 mammalian cells resulted in ADE induction, increasing viral infection. This study aimed to express the D54 monoclonal antibody in Nicotiana benthamiana. The plant-produced antibody had a similar neutralizing profile to the previous 54_hG1 antibody. Notably, the ADE activities of the plant-derived antibody were successfully eliminated, with no sign of viral induction. These findings suggest that N. benthamiana could be a source of therapeutic DENV antibodies. The method offers several advantages, including lower ADE, cost-effectiveness, simple facility requirements, scalability, and potential industrial-scale production in GMP facilities.
Collapse
Affiliation(s)
- Supaluk Krittanai
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | | | | | - Pannamthip Pitaksajjakul
- Center of Excellence for Antibody Research, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Sujitra Keadsanti
- Center of Excellence for Antibody Research, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Pongrama Ramasoota
- Center of Excellence for Antibody Research, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Richard Strasser
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190 Vienna, Austria
| | - Waranyoo Phoolcharoen
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
3
|
Singh R, Chandley P, Rohatgi S. Recent Advances in the Development of Monoclonal Antibodies and Next-Generation Antibodies. Immunohorizons 2023; 7:886-897. [PMID: 38149884 PMCID: PMC10759153 DOI: 10.4049/immunohorizons.2300102] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/07/2023] [Indexed: 12/28/2023] Open
Abstract
mAbs are highly indispensable tools for diagnostic, prophylactic, and therapeutic applications. The first technique, hybridoma technology, was based on fusion of B lymphocytes with myeloma cells, which resulted in generation of single mAbs against a specific Ag. Along with hybridoma technology, several novel and alternative methods have been developed to improve mAb generation, ranging from electrofusion to the discovery of completely novel technologies such as B cell immortalization; phage, yeast, bacterial, ribosome, and mammalian display systems; DNA/RNA encoded Abs; single B cell technology; transgenic animals; and artificial intelligence/machine learning. This commentary outlines the evolution, methodology, advantages, and limitations of various mAb production techniques. Furthermore, with the advent of next-generation Ab technologies such as single-chain variable fragments, nanobodies, bispecific Abs, Fc-engineered Abs, Ab biosimilars, Ab mimetics, and Ab-drug conjugates, the healthcare and pharmaceutical sectors have become resourceful to develop highly specific mAb treatments against various diseases such as cancer and autoimmune and infectious diseases.
Collapse
Affiliation(s)
- Rohit Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand, India
| | - Pankaj Chandley
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand, India
| | - Soma Rohatgi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand, India
| |
Collapse
|
4
|
Balinsky CA, Jiang L, Jani V, Cheng Y, Zhang Z, Belinskaya T, Qiu Q, Long TK, Schilling MA, Jenkins SA, Corson KS, Martin NJ, Letizia AG, Hontz RD, Sun P. Antibodies to S2 domain of SARS-CoV-2 spike protein in Moderna mRNA vaccinated subjects sustain antibody-dependent NK cell-mediated cell cytotoxicity against Omicron BA.1. Front Immunol 2023; 14:1266829. [PMID: 38077368 PMCID: PMC10702584 DOI: 10.3389/fimmu.2023.1266829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/24/2023] [Indexed: 12/18/2023] Open
Abstract
Vaccination with the primary two-dose series of SARS-CoV-2 mRNA protects against infection with the ancestral strain, and limits the presentation of severe disease after re-infection by multiple variants of concern (VOC), including Omicron, despite the lack of a strong neutralizing response to these variants. We compared antibody responses in serum samples collected from mRNA-1273 (Moderna) vaccinated subjects to identify mechanisms of immune escape and cross-protection. Using pseudovirus constructs containing domain-specific amino acid changes representative of Omicron BA.1, combined with domain competition and RBD-antibody depletion, we showed that RBD antibodies were primarily responsible for virus neutralization and variant escape. Antibodies to NTD played a less significant role in antibody neutralization but acted along with RBD to enhance neutralization. S2 of Omicron BA.1 had no impact on neutralization escape, suggesting it is a less critical domain for antibody neutralization; however, it was as capable as S1 at eliciting IgG3 responses and NK-cell mediated, antibody-dependent cell cytotoxicity (ADCC). Antibody neutralization and ADCC activities to RBD, NTD, and S1 were all prone to BA.1 escape. In contrast, ADCC activities to S2 resisted BA.1 escape. In conclusion, S2 antibodies showed potent ADCC function and resisted Omicron BA.1 escape, suggesting that S2 contributes to cross-protection against Omicron BA.1. In line with its conserved nature, S2 may hold promise as a vaccine target against future variants of SARS-CoV-2.
Collapse
Affiliation(s)
- Corey A. Balinsky
- Henry Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Le Jiang
- Henry Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Vihasi Jani
- Henry Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | | | - Zhiwen Zhang
- Henry Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Tatyana Belinskaya
- Henry Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Qi Qiu
- Henry Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | | | - Megan A. Schilling
- Virology and Emerging Infectious Department, U.S. Naval Medical Research Unit SOUTH, Lima, Peru
| | - Sarah A. Jenkins
- Diagnostics and Surveillance Department, Naval Medical Research Command, Silver Spring, MD, United States
| | - Karen S. Corson
- US Naval Medical Research Unit-INDO PACIFIC, Singapore, Singapore
| | | | | | - Robert D. Hontz
- US Naval Medical Research Unit-INDO PACIFIC, Singapore, Singapore
| | - Peifang Sun
- Diagnostics and Surveillance Department, Naval Medical Research Command, Silver Spring, MD, United States
| |
Collapse
|
5
|
Sun H, Yang M, Lai H, Neupane B, Teh AYH, Jugler C, Ma JKC, Steinkellner H, Bai F, Chen Q. A Dual-Approach Strategy to Optimize the Safety and Efficacy of Anti-Zika Virus Monoclonal Antibody Therapeutics. Viruses 2023; 15:1156. [PMID: 37243242 PMCID: PMC10221487 DOI: 10.3390/v15051156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Antibody-dependent enhancement of infection (ADE) is clinically relevant to Dengue virus (DENV) infection and poses a major risk to the application of monoclonal antibody (mAb)-based therapeutics against related flaviviruses such as the Zika virus (ZIKV). Here, we tested a two-tier approach for selecting non-cross-reactive mAbs combined with modulating Fc glycosylation as a strategy to doubly secure the elimination of ADE while preserving Fc effector functions. To this end, we selected a ZIKV-specific mAb (ZV54) and generated three ZV54 variants using Chinese hamster ovary cells and wild-type (WT) and glycoengineered ΔXF Nicotiana benthamiana plants as production hosts (ZV54CHO, ZV54WT, and ZV54ΔXF). The three ZV54 variants shared an identical polypeptide backbone, but each exhibited a distinct Fc N-glycosylation profile. All three ZV54 variants showed similar neutralization potency against ZIKV but no ADE activity for DENV infection, validating the importance of selecting the virus/serotype-specific mAbs for avoiding ADE by related flaviviruses. For ZIKV infection, however, ZV54CHO and ZV54ΔXF showed significant ADE activity while ZV54WT completely forwent ADE, suggesting that Fc glycan modulation may yield mAb glycoforms that abrogate ADE even for homologous viruses. In contrast to the current strategies for Fc mutations that abrogate all effector functions along with ADE, our approach allowed the preservation of effector functions as all ZV54 glycovariants retained antibody-dependent cellular cytotoxicity (ADCC) against the ZIKV-infected cells. Furthermore, the ADE-free ZV54WT demonstrated in vivo efficacy in a ZIKV-infection mouse model. Collectively, our study provides further support for the hypothesis that antibody-viral surface antigen and Fc-mediated host cell interactions are both prerequisites for ADE, and that a dual-approach strategy, as shown herein, contributes to the development of highly safe and efficacious anti-ZIKV mAb therapeutics. Our findings may be impactful to other ADE-prone viruses, including SARS-CoV-2.
Collapse
Affiliation(s)
- Haiyan Sun
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Ming Yang
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Huafang Lai
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Biswas Neupane
- Department of Cell and Molecular Biology, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Audrey Y.-H. Teh
- Institute for Infection and Immunity, St. George’s, University of London, London SW17 0RE, UK
| | - Collin Jugler
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Julian K.-C. Ma
- Institute for Infection and Immunity, St. George’s, University of London, London SW17 0RE, UK
| | - Herta Steinkellner
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, 1180 Vienna, Austria
| | - Fengwei Bai
- Department of Cell and Molecular Biology, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Qiang Chen
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
6
|
Yang M, Sun H, Lai H, Neupane B, Bai F, Steinkellner H, Chen Q. Plant-Produced Anti-Zika Virus Monoclonal Antibody Glycovariant Exhibits Abrogated Antibody-Dependent Enhancement of Infection. Vaccines (Basel) 2023; 11:755. [PMID: 37112665 PMCID: PMC10144123 DOI: 10.3390/vaccines11040755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023] Open
Abstract
Monoclonal antibodies (mAb) against the envelope (E) protein of Zika virus (ZIKV) have shown great potential as therapeutics against the Zika epidemics. However, their use as a therapy may predispose treated individuals to severe infection by the related dengue virus (DENV) via antibody-dependent enhancement of infection (ADE). Here, we generated a broadly neutralizing flavivirus mAb, ZV1, with an identical protein backbone but different Fc glycosylation profiles. The three glycovariants, produced in wild-type (WT) and glycoengineered ΔXF Nicotiana benthamiana plants and in Chinese hamster ovary cells (ZV1WT, ZV1ΔXF, and ZV1CHO), respectively, showed equivalent neutralization potency against both ZIKV and DENV. By contrast, the three mAb glycoforms demonstrated drastically different ADE activity for DENV and ZIKV infection. While ZV1CHO and ZV1ΔXF showed ADE activity upon DENV and ZIKV infection, ZV1WT totally forwent its ADE. Importantly, all three glycovariants exhibited antibody-dependent cellular cytotoxicity (ADCC) against virus-infected cells, with increased potency by the fucose-free ZV1ΔXF glycoform. Moreover, the in vivo efficacy of the ADE-free ZV1WT was demonstrated in a murine model. Collectively, we demonstrated the feasibility of modulating ADE by Fc glycosylation, thereby establishing a novel approach for improving the safety of flavivirus therapeutics. Our study also underscores the versatile use of plants for the rapid expression of complex human proteins to reveal novel insight into antibody function and viral pathogenesis.
Collapse
Affiliation(s)
- Ming Yang
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ 85225, USA
| | - Haiyan Sun
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ 85225, USA
| | - Huafang Lai
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ 85225, USA
| | - Biswas Neupane
- Department of Cell and Molecular Biology, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Fengwei Bai
- Department of Cell and Molecular Biology, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Herta Steinkellner
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, 1180 Vienna, Austria
| | - Qiang Chen
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ 85225, USA
| |
Collapse
|
7
|
Machain-Williams C, Reyes-Solis GC, Blitvich BJ, Laredo-Tiscareño V, Dzul-Rosado AR, Kim S, AbuBakar S. Evaluation of an Immunoglobulin E Capture Enzyme-Linked Immunosorbent Assay for the Early Diagnosis of Dengue. Viral Immunol 2023; 36:101-109. [PMID: 36862827 DOI: 10.1089/vim.2022.0110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
Dengue virus (DENV) is the etiological agent of dengue, the most important mosquito-transmitted viral disease of humans worldwide. Enzyme-linked immunosorbent assays (ELISAs) designed to detect DENV IgM are commonly used for dengue diagnosis. However, DENV IgM is not reliably detected until ≥4 days after illness onset. Reverse transcription-polymerase chain reaction (RT-PCR) can diagnose early dengue but requires specialized equipment, reagents, and trained personnel. Additional diagnostic tools are needed. Limited work has been performed to determine whether IgE-based assays can be used for the early detection of vector-borne viral diseases, including dengue. In this study, we determined the efficacy of a DENV IgE capture ELISA for the detection of early dengue. Sera were collected within the first 4 days of illness onset from 117 patients with laboratory-confirmed dengue, as determined by DENV-specific RT-PCR. The serotypes responsible for the infections were DENV-1 and DENV-2 (57 and 60 patients, respectively). Sera were also collected from 113 dengue-negative individuals with febrile illness of undetermined etiology and 30 healthy controls. The capture ELISA detected DENV IgE in 97 (82.9%) confirmed dengue patients and none of the healthy controls. There was a high false positivity rate (22.1%) among the febrile non-dengue patients. In conclusion, we provide evidence that IgE capture assays have the potential to be explored for early diagnosis of dengue, but further research is necessary to address the possible false positivity rate among patients with other febrile illnesses.
Collapse
Affiliation(s)
- Carlos Machain-Williams
- Laboratorio de Arbovirologia, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi," Universidad Autónoma de Yucatan, Merida, Yucatan, Mexico
| | - Guadalupe C Reyes-Solis
- Laboratorio de Arbovirologia, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi," Universidad Autónoma de Yucatan, Merida, Yucatan, Mexico
| | - Bradley J Blitvich
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - Viridiana Laredo-Tiscareño
- Laboratorio de Arbovirologia, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi," Universidad Autónoma de Yucatan, Merida, Yucatan, Mexico.,Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | | | - Sungmin Kim
- Department of Infectious Diseases in Internal Medicine, Sejong Chungnam National University Hospital, School of Medicine, Chungnam National University, Sejong, Korea
| | - Sazaly AbuBakar
- Tropical Infectious Diseases Research and Education Centre (TIDREC), Higher Institution Center of Excellence (HICOE), Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
8
|
Teo A, Tan HD, Loy T, Chia PY, Chua CLL. Understanding antibody-dependent enhancement in dengue: Are afucosylated IgG1s a concern? PLoS Pathog 2023; 19:e1011223. [PMID: 36996026 PMCID: PMC10062565 DOI: 10.1371/journal.ppat.1011223] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
Affiliation(s)
- Andrew Teo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Medicine, The Doherty Institute, University of Melbourne, Melbourne, Australia
| | - Hao Dong Tan
- School of Biosciences, Faculty of Health and Medicine Sciences, Taylor’s University, Subang Jaya, Malaysia
| | - Thomas Loy
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Po Ying Chia
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Caroline Lin Lin Chua
- School of Biosciences, Faculty of Health and Medicine Sciences, Taylor’s University, Subang Jaya, Malaysia
| |
Collapse
|
9
|
Immune-Mediated Pathogenesis in Dengue Virus Infection. Viruses 2022; 14:v14112575. [PMID: 36423184 PMCID: PMC9699586 DOI: 10.3390/v14112575] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022] Open
Abstract
Dengue virus (DENV) infection is one of the major public health concerns around the globe, especially in the tropical regions of the world that contribute to 75% percent of dengue cases. While the majority of DENV infections are mild or asymptomatic, approximately 5% of the cases develop a severe form of the disease that is mainly attributed to sequential infection with different DENV serotypes. The severity of dengue depends on many immunopathogenic mechanisms involving both viral and host factors. Emerging evidence implicates an impaired immune response as contributing to disease progression and severity by restricting viral clearance and inducing severe inflammation, subsequently leading to dengue hemorrhagic fever and dengue shock syndrome. Moreover, the ability of DENV to infect a wide variety of immune cells, including monocytes, macrophages, dendritic cells, mast cells, and T and B cells, further dysregulates the antiviral functions of these cells, resulting in viral dissemination. Although several risk factors associated with disease progression have been proposed, gaps persist in the understanding of the disease pathogenesis and further investigations are warranted. In this review, we discuss known mechanisms of DENV-mediated immunopathogenesis and its association with disease progression and severity.
Collapse
|
10
|
SARS-CoV-2 Antinucleocapsid Antibody Response of mRNA and Inactivated Virus Vaccines Compared to Unvaccinated Individuals. Vaccines (Basel) 2022; 10:vaccines10050643. [PMID: 35632399 PMCID: PMC9143597 DOI: 10.3390/vaccines10050643] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/07/2022] [Accepted: 04/14/2022] [Indexed: 11/29/2022] Open
Abstract
Comparative studies of SARS-CoV-2 antinucleocapsid (anti-N) antibody response in the context of inactivated virus vaccines versus natural infection are limited. This study aims to determine and compare the anti-N antibody levels in people vaccinated with Sinopharm’s (Wuhan, China) inactivated virus vaccine in comparison with naturally infected unvaccinated and Pfizer’s spike (S) mRNA-based vaccinated subjects. Two hundred ninety-nine Jordanian adults participated in the study including unvaccinated COVID-19-infected patients (n = 99), Pfizer-vaccinated (n = 100), and Sinopharm-vaccinated recipients (n = 100). Serum samples were assayed for anti-N IgG, anti-N IgM, and anti-S IgG. Sera of 64.6% of naturally infected unvaccinated participants had positive anti-S IgG (median = 36.35 U/mL; range: 0.04−532.5 U/mL) compared to 88% of Pfizer-vaccinated (Manhattan, NY, USA) (median = 26.52 U/mL; range: 0.39−1265 U/mL) and 58% of Sinopharm-vaccinated subjects (median = 14.35 U/mL; range: 0.39−870.17 U/mL). Samples of 60.6% of naturally infected unvaccinated people had positive anti-N IgG (median = 15.03 U/mL; range: 0−265.1 U/mL) compared to 25% of Pfizer-vaccinated (median = 0.02 U/mL; range: 0−68 U/mL) and 48% of Sinopharm-vaccinated subjects (median = 0.8 U/mL; range: 0−146.3 U/mL). Anti-N titers among the three groups were significantly different (p < 0.05). Anti-N IgM antibodies appeared in 23.2% of the naturally infected unvaccinated group (median = 0.29 U/mL; range: 0−15 U/mL) compared to only 9.0% of Pfizer-vaccinated (median = 018 U/mL; range: 0−33 U/mL) and 7.0% of Sinopharm-vaccinated subjects (median = 0.2 U/mL; range: 0−12.02 U/mL). A significant negative correlation was found between anti-S and age for both vaccines and between anti-S and the presence of chronic disease in Sinopharm-vaccinated subjects. A significant positive correlation between anti-N and anti-S titers was found among the three groups. This study shows that the inactivated virus vaccine, Sinopharm, induces an anti-N response that can boost that of natural infection or vice versa. On the other hand, the Pfizer mRNA-based vaccine induces a significantly stronger anti-S Ab response.
Collapse
|
11
|
Tien SM, Chang PC, Lai YC, Chuang YC, Tseng CK, Kao YS, Huang HJ, Hsiao YP, Liu YL, Lin HH, Chu CC, Cheng MH, Ho TS, Chang CP, Ko SF, Shen CP, Anderson R, Lin YS, Wan SW, Yeh TM. Therapeutic efficacy of humanized monoclonal antibodies targeting dengue virus nonstructural protein 1 in the mouse model. PLoS Pathog 2022; 18:e1010469. [PMID: 35486576 PMCID: PMC9053773 DOI: 10.1371/journal.ppat.1010469] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 03/24/2022] [Indexed: 12/27/2022] Open
Abstract
Dengue virus (DENV) which infects about 390 million people per year in tropical and subtropical areas manifests various disease symptoms, ranging from fever to life-threatening hemorrhage and even shock. To date, there is still no effective treatment for DENV disease, but only supportive care. DENV nonstructural protein 1 (NS1) has been shown to play a key role in disease pathogenesis. Recent studies have shown that anti-DENV NS1 antibody can provide disease protection by blocking the DENV-induced disruption of endothelial integrity. We previously demonstrated that anti-NS1 monoclonal antibody (mAb) protected mice from all four serotypes of DENV challenge. Here, we generated humanized anti-NS1 mAbs and transferred them to mice after DENV infection. The results showed that DENV-induced prolonged bleeding time and skin hemorrhage were reduced, even several days after DENV challenge. Mechanistic studies showed the ability of humanized anti-NS1 mAbs to inhibit NS1-induced vascular hyperpermeability and to elicit Fcγ-dependent complement-mediated cytolysis as well as antibody-dependent cellular cytotoxicity of cells infected with four serotypes of DENV. These results highlight humanized anti-NS1 mAb as a potential therapeutic agent in DENV infection. DENV comprising four serotypes has a complicated pathogenesis and remains an unresolved global health problem. To date, supportive therapy is the mainstay for treatment of dengue patients. Despite a licensed Sanofi vaccine and ongoing clinical trials, more effective vaccines and/or licensed therapeutic drugs are required. Therapeutic mAbs are a potential tool to treat many epidemic diseases because of their high target specificity. Humanized anti-NS1 mAbs can recognize the NS1 from all four serotypes of DENV without danger of inducing ADE. In the DENV infection mouse model, we demonstrate that humanized NS1 mAbs have therapeutic benefits such as reducing DENV-induced prolonged bleeding time and skin hemorrhage. In vitro mechanistic studies showed a reduction of NS1-induced vascular permeability and an increase in cytolysis of DENV-infected cells. Our results showed that humanized anti-NS1 mAbs show strong potential for development toward clinical use.
Collapse
Affiliation(s)
- Sen-Mao Tien
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Po-Chun Chang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Leadgene Biomedical, Inc. Tainan, Taiwan
| | - Yen-Chung Lai
- Leadgene Biomedical, Inc. Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yung-Chun Chuang
- Leadgene Biomedical, Inc. Tainan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | - Yu-San Kao
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hong-Jyun Huang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Peng Hsiao
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ling Liu
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsing-Han Lin
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- SIDSCO Biomedical Co., Ltd. Kaohsiung, Taiwan
| | - Chien-Chou Chu
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Miao-Huei Cheng
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzong-Shiann Ho
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Peng Chang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Shu-Fen Ko
- Development Center for Biotechnology, Taipei, Taiwan
| | - Che-Piao Shen
- Development Center for Biotechnology, Taipei, Taiwan
| | - Robert Anderson
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Canada
| | - Yee-Shin Lin
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
- * E-mail: (YSL); (SWW); (TMY)
| | - Shu-Wen Wan
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
- * E-mail: (YSL); (SWW); (TMY)
| | - Trai-Ming Yeh
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
- * E-mail: (YSL); (SWW); (TMY)
| |
Collapse
|
12
|
Gunn BM, Lu R, Slein MD, Ilinykh PA, Huang K, Atyeo C, Schendel SL, Kim J, Cain C, Roy V, Suscovich TJ, Takada A, Halfmann PJ, Kawaoka Y, Pauthner MG, Momoh M, Goba A, Kanneh L, Andersen KG, Schieffelin JS, Grant D, Garry RF, Saphire EO, Bukreyev A, Alter G. A Fc engineering approach to define functional humoral correlates of immunity against Ebola virus. Immunity 2021; 54:815-828.e5. [PMID: 33852832 PMCID: PMC8111768 DOI: 10.1016/j.immuni.2021.03.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 01/28/2021] [Accepted: 03/16/2021] [Indexed: 01/31/2023]
Abstract
Protective Ebola virus (EBOV) antibodies have neutralizing activity and induction of antibody constant domain (Fc)-mediated innate immune effector functions. Efforts to enhance Fc effector functionality often focus on maximizing antibody-dependent cellular cytotoxicity, yet distinct combinations of functions could be critical for antibody-mediated protection. As neutralizing antibodies have been cloned from EBOV disease survivors, we sought to identify survivor Fc effector profiles to help guide Fc optimization strategies. Survivors developed a range of functional antibody responses, and we therefore applied a rapid, high-throughput Fc engineering platform to define the most protective profiles. We generated a library of Fc variants with identical antigen-binding fragments (Fabs) from an EBOV neutralizing antibody. Fc variants with antibody-mediated complement deposition and moderate natural killer (NK) cell activity demonstrated complete protective activity in a stringent in vivo mouse model. Our findings highlight the importance of specific effector functions in antibody-mediated protection, and the experimental platform presents a generalizable resource for identifying correlates of immunity to guide therapeutic antibody design.
Collapse
Affiliation(s)
- Bronwyn M Gunn
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Richard Lu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Matthew D Slein
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Philipp A Ilinykh
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, USA; Galveston National Laboratory, Galveston, TX, USA
| | - Kai Huang
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, USA; Galveston National Laboratory, Galveston, TX, USA
| | - Caroline Atyeo
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | | | - Jiyoung Kim
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Caitlin Cain
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Vicky Roy
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | | | - Ayato Takada
- Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Peter J Halfmann
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Matthias G Pauthner
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA
| | - Mambu Momoh
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, Kenema, Sierra Leone
| | - Augustine Goba
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, Kenema, Sierra Leone
| | - Lansana Kanneh
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, Kenema, Sierra Leone
| | - Kristian G Andersen
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA; Scripps Research Translational Institute, La Jolla, CA, USA
| | - John S Schieffelin
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA, USA
| | - Donald Grant
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, Kenema, Sierra Leone; Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - Robert F Garry
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, USA; Galveston National Laboratory, Galveston, TX, USA; Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX, USA.
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
13
|
Batra M, Tian R, Zhang C, Clarence E, Sacher CS, Miranda JN, De La Fuente JRO, Mathew M, Green D, Patel S, Bastidas MVP, Haddadi S, Murthi M, Gonzalez MS, Kambali S, Santos KHM, Asif H, Modarresi F, Faghihi M, Mirsaeidi M. Role of IgG against N-protein of SARS-CoV2 in COVID19 clinical outcomes. Sci Rep 2021; 11:3455. [PMID: 33568776 PMCID: PMC7875990 DOI: 10.1038/s41598-021-83108-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/29/2021] [Indexed: 02/08/2023] Open
Abstract
The Nucleocapsid Protein (N Protein) of severe acute respiratory syndrome Coronavirus 2 (SARS-CoV2) is located in the viral core. Immunoglobulin G (IgG) targeting N protein is detectable in the serum of infected patients. The effect of high titers of IgG against N-protein on clinical outcomes of SARS-CoV2 disease has not been described. We studied 400 RT-PCR confirmed SARS-CoV2 patients to determine independent factors associated with poor outcomes, including Medical Intensive Care Unit (MICU) admission, prolonged MICU stay and hospital admissions, and in-hospital mortality. We also measured serum IgG against the N protein and correlated its concentrations with clinical outcomes. We found that several factors, including Charlson comorbidity Index (CCI), high levels of IL6, and presentation with dyspnea were associated with poor clinical outcomes. It was shown that higher CCI and higher IL6 levels were independently associated with in-hospital mortality. Anti-N protein IgG was detected in the serum of 55 (55%) patients at the time of admission. A high concentration of antibodies, defined as signal to cut off ratio (S/Co) > 1.5 (75 percentile of all measurements), was found in 25 (25%) patients. The multivariable logistic regression models showed that between being an African American, higher CCI, lymphocyte counts, and S/Co ratio > 1.5, only S/Co ratio were independently associated with MICU admission and longer length of stay in hospital. This study recommends that titers of IgG targeting N-protein of SARS-CoV2 at admission is a prognostic factor for the clinical course of disease and should be measured in all patients with SARS-CoV2 infection.
Collapse
Affiliation(s)
- Mayank Batra
- Division of Pulmonary and Critical Care, University of Miami, 1600 NW 10th Ave # 7072B, Miami, FL, 33136, USA
| | - Runxia Tian
- Division of Pulmonary and Critical Care, University of Miami, 1600 NW 10th Ave # 7072B, Miami, FL, 33136, USA
| | - Chongxu Zhang
- Division of Pulmonary and Critical Care, University of Miami, 1600 NW 10th Ave # 7072B, Miami, FL, 33136, USA
| | | | | | | | | | - Megan Mathew
- School of Medicine, University of Miami, Miami, FL, USA
| | - Desmond Green
- School of Medicine, University of Miami, Miami, FL, USA
| | - Sayari Patel
- School of Medicine, University of Miami, Miami, FL, USA
| | | | - Sara Haddadi
- Division of Pulmonary and Critical Care, University of Miami, 1600 NW 10th Ave # 7072B, Miami, FL, 33136, USA
| | - Mukunthan Murthi
- Division of Pulmonary and Critical Care, University of Miami, 1600 NW 10th Ave # 7072B, Miami, FL, 33136, USA
| | - Miguel Santiago Gonzalez
- Division of Pulmonary and Critical Care, University of Miami, 1600 NW 10th Ave # 7072B, Miami, FL, 33136, USA
| | - Shweta Kambali
- Division of Pulmonary and Critical Care, University of Miami, 1600 NW 10th Ave # 7072B, Miami, FL, 33136, USA
| | - Kayo H M Santos
- Division of Pulmonary and Critical Care, University of Miami, 1600 NW 10th Ave # 7072B, Miami, FL, 33136, USA
| | - Huda Asif
- Division of Pulmonary and Critical Care, University of Miami, 1600 NW 10th Ave # 7072B, Miami, FL, 33136, USA
| | | | | | - Mehdi Mirsaeidi
- Division of Pulmonary and Critical Care, University of Miami, 1600 NW 10th Ave # 7072B, Miami, FL, 33136, USA.
| |
Collapse
|
14
|
Huang K, Lin M, Kuo T, Chen C, Lin C, Chou Y, Chao T, Pang Y, Kao H, Huang R, Lin S, Chang S, Yang P. Humanized COVID-19 decoy antibody effectively blocks viral entry and prevents SARS-CoV-2 infection. EMBO Mol Med 2021; 13:e12828. [PMID: 33159417 PMCID: PMC7799362 DOI: 10.15252/emmm.202012828] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 12/17/2022] Open
Abstract
To circumvent the devastating pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, a humanized decoy antibody (ACE2-Fc fusion protein) was designed to target the interaction between viral spike protein and its cellular receptor, angiotensin-converting enzyme 2 (ACE2). First, we demonstrated that ACE2-Fc could specifically abrogate virus replication by blocking the entry of SARS-CoV-2 spike-expressing pseudotyped virus into both ACE2-expressing lung cells and lung organoids. The impairment of viral entry was not affected by virus variants, since efficient inhibition was also observed in six SARS-CoV-2 clinical strains, including the D614G variants which have been shown to exhibit increased infectivity. The preservation of peptidase activity also enables ACE2-Fc to reduce the angiotensin II-mediated cytokine cascade. Furthermore, this Fc domain of ACE2-Fc was shown to activate NK cell degranulation after co-incubation with Spike-expressing H1975 cells. These promising characteristics potentiate the therapeutic prospects of ACE2-Fc as an effective treatment for COVID-19.
Collapse
Affiliation(s)
- Kuo‐Yen Huang
- Institute of Biomedical SciencesAcademia SinicaTaipeiTaiwan
| | - Ming‐Shiu Lin
- Institute of Biomedical SciencesAcademia SinicaTaipeiTaiwan
| | - Ting‐Chun Kuo
- Department of Internal MedicineNational Taiwan University Hospital and National Taiwan University College of MedicineTaipeiTaiwan
| | - Ci‐Ling Chen
- Institute of Biomedical SciencesAcademia SinicaTaipeiTaiwan
| | - Chung‐Chih Lin
- Institute of Biomedical SciencesAcademia SinicaTaipeiTaiwan
| | - Yu‐Chi Chou
- Biomedical Translation Research Center (BioTReC)Academia SinicaTaipeiTaiwan
| | - Tai‐Ling Chao
- Department of Clinical Laboratory Sciences and Medical BiotechnologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Yu‐Hao Pang
- Department of Clinical Laboratory Sciences and Medical BiotechnologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Han‐Chieh Kao
- Department of Clinical Laboratory Sciences and Medical BiotechnologyNational Taiwan University College of MedicineTaipeiTaiwan
| | - Rih‐Sheng Huang
- Institute of Biological ChemistryAcademia SinicaTaipeiTaiwan
| | - Steven Lin
- Institute of Biological ChemistryAcademia SinicaTaipeiTaiwan
- Institute of Biochemical SciencesNational Taiwan UniversityTaipeiTaiwan
| | - Sui‐Yuan Chang
- Department of Clinical Laboratory Sciences and Medical BiotechnologyNational Taiwan University College of MedicineTaipeiTaiwan
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Pan‐Chyr Yang
- Institute of Biomedical SciencesAcademia SinicaTaipeiTaiwan
- Department of Internal MedicineNational Taiwan University Hospital and National Taiwan University College of MedicineTaipeiTaiwan
- Genomics Research CenterAcademia SinicaTaipeiTaiwan
| |
Collapse
|
15
|
Liao YH, Hsu RJ, Wang TH, Wu CT, Huang SY, Hsu CY, Su YC, Hsu WL, Liu DW. Aspirin decreases hepatocellular carcinoma risk in hepatitis C virus carriers: a nationwide cohort study. BMC Gastroenterol 2020; 20:6. [PMID: 31918672 PMCID: PMC6953130 DOI: 10.1186/s12876-020-1158-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 12/31/2019] [Indexed: 12/21/2022] Open
Abstract
Background Aspirin has been found to lower the occurrence rates of some cancers through the inhibition of the cyclooxygenase enzyme. For example, there is a well-known association between aspirin use and the occurrence of hepatocellular carcinoma (HCC) in hepatitis B virus (HBV) carriers. However, the association, if any, between aspirin use and HCC in hepatitis C virus (HCV) carriers is unknown. Therefore, this study compared the occurrence rates of HCC in HCV carriers treated with or without aspirin. Methods The participants in this retrospective cohort study consisted of people newly diagnosed with HCV in Taiwan from 2000 to 2012. Those who were treated with aspirin were defined as the control group, whereas those not treated with aspirin were defined as the comparison cohort. We used a 1:1 propensity score matching by age, sex, comorbidities, drugs, diagnosis year, and index year with covariate assessment. Results Our study sample consisted of 2980 aspirin-treated HCV carriers and 7771 non-aspirin-treated HCV carriers. After propensity score matching, each cohort consisted of 1911 HCV carriers. The adjusted hazard ratio (aHR) of HCC incidence in the aspirin users (aHR = 0.56, 95% CI = 0.43–0.72, p < 0.001) was significantly lower than that in the non-aspirin users. A Kaplan-Meier analysis showed that among the HCV carriers, the aspirin users had a lower cumulative incidence rate of HCC over the first 10 years of aspirin treatment (p < 0.0001). Conclusions The HCC incidence rate was lower in the aspirin-using HCV carriers than in the non- aspirin-using HCV carriers, indicating that the effects of aspirin might occur through inhibition of the cyclooxygenase enzyme pathway. Moreover, protection from HCC was provided by less than a year of aspirin treatment, while treatment with aspirin for 1 to 2 years exhibited the greatest protective effect. We therefore encourage aspirin treatment to prevent HCC in HCV carriers.
Collapse
Affiliation(s)
- Yen-Hsiang Liao
- Department of Radiation Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Ren-Jun Hsu
- Department of Radiation Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Tzu-Hwei Wang
- Department of Radiation Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chen-Ta Wu
- Department of Radiation Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Sheng-Yao Huang
- Department of Radiation Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chung-Y Hsu
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Yuan-Chih Su
- College of Medicine, China Medical University, Taichung, Taiwan.,Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Lin Hsu
- Department of Radiation Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Dai-Wei Liu
- Department of Radiation Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan. .,School of Medicine, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
16
|
Gyurova IE, Ali A, Waggoner SN. Natural Killer Cell Regulation of B Cell Responses in the Context of Viral Infection. Viral Immunol 2019; 33:334-341. [PMID: 31800366 DOI: 10.1089/vim.2019.0129] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Secretion of both neutralizing and nonneutralizing virus-specific antibodies by B cells is a key component of immune control of many virus infections and a critical benchmark of successful preventative vaccines. Natural killer (NK) cells also play a vital role in antiviral immune defense via cytolytic elimination of infected cells and production of proinflammatory antiviral cytokines. Accumulating evidence points to multifaceted crosstalk between NK cells and antiviral B cell responses that can determine virus elimination, pathogenesis of infection, and efficacy of vaccine-elicited protection. These outcomes are a result of both positive and negative influences of NK cells on the B cell responses, as well as canonical antiviral killing of infected B cells. On one hand, NK cell-derived cytokines such as interferon-gamma (IFN-γ) may promote B cell activation and enhance immunoglobulin production. In contrast, NK cell immunoregulatory killing of CD4 T cells can limit affinity maturation in germinal centers resulting in weak infection or vaccine induction of antiviral neutralizing antibodies. In this review, we will discuss these and other dueling contributions of NK cells to B cell responses during virus infection or vaccination.
Collapse
Affiliation(s)
- Ivayla E Gyurova
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Pathobiology and Molecular Medicine Graduate Program, University of Cincinnati, Cincinnati, Ohio, USA
| | - Ayad Ali
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Medical Scientist Training Program, University of Cincinnati, Cincinnati, Ohio, USA.,Graduate Program in Immunology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Stephen N Waggoner
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Pathobiology and Molecular Medicine Graduate Program, University of Cincinnati, Cincinnati, Ohio, USA.,Medical Scientist Training Program, University of Cincinnati, Cincinnati, Ohio, USA.,Graduate Program in Immunology, University of Cincinnati, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
17
|
Petitdemange C, Maucourant C, Tarantino N, Rey J, Vieillard V. Glycogen synthetase kinase 3 inhibition drives MIC-A/B to promote cytokine production by human natural killer cells in Dengue virus type 2 infection. Eur J Immunol 2019; 50:342-352. [PMID: 31743425 DOI: 10.1002/eji.201948284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/11/2019] [Accepted: 11/15/2019] [Indexed: 01/23/2023]
Abstract
Dengue virus (DENV) is the most widespread arbovirus worldwide and is responsible for major outbreaks. The host's immune response plays a crucial role in controlling this infection but might also contribute to the promotion of viral spread and immunopathology. In response to DENV infection, NK cells preferentially produce cytokines and are cytotoxic in the presence of specific antibodies. Here, we identified that DENV-2 inhibits glycogen synthase kinase 3 (GSK-3) activity to subsequently induce MHC class-1-related chain (MIC) A and MIC-B expression and IL-12 production in monocyte-derived DCs, independently of the STAT-3 pathway. The inhibition of GSK-3 by DENV-2 or small molecules induced MIC-A/B expression on monocyte-derived DCs, resulting in autologous NK cells of a specific increase in IFN-γ and TNF-α production, in the absence of direct cytotoxicity. Together, these findings identified GSK-3 as a regulator of MIC-A/B expression and suggested its role in DENV-2 infection to specifically induce cytokine production by NK cells.
Collapse
Affiliation(s)
- Caroline Petitdemange
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, UPMC, Paris, France
| | - Christopher Maucourant
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, UPMC, Paris, France
| | - Nadine Tarantino
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, UPMC, Paris, France
| | - Juliana Rey
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, UPMC, Paris, France
| | - Vincent Vieillard
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, UPMC, Paris, France
| |
Collapse
|
18
|
McKechnie JL, Beltrán D, Pitti A, Saenz L, Araúz AB, Vergara R, Harris E, Lanier LL, Blish CA, López-Vergès S. HLA Upregulation During Dengue Virus Infection Suppresses the Natural Killer Cell Response. Front Cell Infect Microbiol 2019; 9:268. [PMID: 31396492 PMCID: PMC6663972 DOI: 10.3389/fcimb.2019.00268] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/10/2019] [Indexed: 11/13/2022] Open
Abstract
Dengue virus (DENV) is the most prevalent mosquito-borne virus in the world and a major cause of morbidity in the tropics and subtropics. Upregulation of HLA class I molecules has long been considered a feature of DENV infection, yet this has not been evaluated in the setting of natural infection. Natural killer (NK) cells, an innate immune cell subset critical for mounting an early response to viral infection, are inhibited by self HLA class I, suggesting that upregulation of HLA class I during DENV infection could dampen the NK cell response. Here we addressed whether upregulation of HLA class I molecules occurs during in vivo DENV infection and, if so, whether this suppresses the NK cell response. We found that HLA class I expression was indeed upregulated during acute DENV infection across multiple cell lineages in vivo. To better understand the role of HLA class I upregulation, we infected primary human monocytes, a major target of DENV infection, in vitro. Upregulation of total HLA class I is dependent on active viral replication and is mediated in part by cytokines and other soluble factors induced by infection, while upregulation of HLA-E occurs in the presence of replication-incompetent virus. Importantly, blocking DENV-infected monocytes with a pan-HLA class I Fab nearly doubles the frequency of degranulating NK cells, while blocking HLA-E does not significantly improve the NK cell response. These findings demonstrate that upregulation of HLA class I during DENV infection suppresses the NK cell response, potentially contributing to disease pathogenesis.
Collapse
Affiliation(s)
- Julia L. McKechnie
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, United States
| | - Davis Beltrán
- Department of Research in Virology and Biotechnology, Gorgas Memorial Institute for Health Studies, Panama City, Panama
- Institute for Scientific Research and Technology Services (INDICASAT-AIP), Panama City, Panama
- Department of Biotechnology, Acharya Nagarjuna University, Guntur, India
| | - Arcelys Pitti
- Department of Research in Virology and Biotechnology, Gorgas Memorial Institute for Health Studies, Panama City, Panama
| | - Lisseth Saenz
- Department of Research in Virology and Biotechnology, Gorgas Memorial Institute for Health Studies, Panama City, Panama
| | | | - Rosemary Vergara
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, United States
| | - Lewis L. Lanier
- Department of Microbiology and Immunology and the Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, United States
| | - Catherine A. Blish
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, United States
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Sandra López-Vergès
- Department of Research in Virology and Biotechnology, Gorgas Memorial Institute for Health Studies, Panama City, Panama
- Institute for Scientific Research and Technology Services (INDICASAT-AIP), Panama City, Panama
| |
Collapse
|