1
|
Watanabe S, Yoshikawa T, Kaku Y, Kurosu T, Fukushi S, Sugimoto S, Nishisaka Y, Fuji H, Marsh G, Maeda K, Ebihara H, Morikawa S, Shimojima M, Saijo M. Construction of a recombinant vaccine expressing Nipah virus glycoprotein using the replicative and highly attenuated vaccinia virus strain LC16m8. PLoS Negl Trop Dis 2023; 17:e0011851. [PMID: 38100536 PMCID: PMC10756534 DOI: 10.1371/journal.pntd.0011851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 12/29/2023] [Accepted: 12/07/2023] [Indexed: 12/17/2023] Open
Abstract
Nipah virus (NiV) is a highly pathogenic zoonotic virus that causes severe encephalitis and respiratory diseases and has a high mortality rate in humans (>40%). Epidemiological studies on various fruit bat species, which are natural reservoirs of the virus, have shown that NiV is widely distributed throughout Southeast Asia. Therefore, there is an urgent need to develop effective NiV vaccines. In this study, we generated recombinant vaccinia viruses expressing the NiV glycoprotein (G) or fusion (F) protein using the LC16m8 strain, and examined their antigenicity and ability to induce immunity. Neutralizing antibodies against NiV were successfully induced in hamsters inoculated with LC16m8 expressing NiV G or F, and the antibody titers were higher than those induced by other vaccinia virus vectors previously reported to prevent lethal NiV infection. These findings indicate that the LC16m8-based vaccine format has superior features as a proliferative vaccine compared with other poxvirus-based vaccines. Moreover, the data collected over the course of antibody elevation during three rounds of vaccination in hamsters provide an important basis for the clinical use of vaccinia virus-based vaccines against NiV disease. Trial Registration: NCT05398796.
Collapse
Affiliation(s)
- Shumpei Watanabe
- Department of Microbiology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Ehime, Japan
- Department of Virology I, National Institute of Infectious Diseases, Musashimurayama, Tokyo, Japan
| | - Tomoki Yoshikawa
- Department of Virology I, National Institute of Infectious Diseases, Musashimurayama, Tokyo, Japan
| | - Yoshihiro Kaku
- Division of Veterinary Science, National Institute of Infectious Diseases, Shinjuku, Tokyo, Japan
| | - Takeshi Kurosu
- Department of Virology I, National Institute of Infectious Diseases, Musashimurayama, Tokyo, Japan
| | - Shuetsu Fukushi
- Department of Virology I, National Institute of Infectious Diseases, Musashimurayama, Tokyo, Japan
| | - Satoko Sugimoto
- Department of Virology I, National Institute of Infectious Diseases, Musashimurayama, Tokyo, Japan
| | - Yuki Nishisaka
- Department of Microbiology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Ehime, Japan
| | - Hikaru Fuji
- Department of Microbiology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Ehime, Japan
| | - Glenn Marsh
- Australian Centre for Disease Preparedness, CSIRO, Geelong, VIC, Australia
| | - Ken Maeda
- Division of Veterinary Science, National Institute of Infectious Diseases, Shinjuku, Tokyo, Japan
| | - Hideki Ebihara
- Department of Virology I, National Institute of Infectious Diseases, Musashimurayama, Tokyo, Japan
| | - Shigeru Morikawa
- Department of Microbiology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Ehime, Japan
| | - Masayuki Shimojima
- Department of Virology I, National Institute of Infectious Diseases, Musashimurayama, Tokyo, Japan
| | - Masayuki Saijo
- Department of Virology I, National Institute of Infectious Diseases, Musashimurayama, Tokyo, Japan
- Public Health Office, Health and Welfare Bureau, Sapporo Municipal Government, Sapporo, Hokkaido, Japan
| |
Collapse
|
2
|
Kitabatake M, Ouji-Sageshima N, Sonobe S, Furukawa R, Konda M, Hara A, Aoki H, Suzuki Y, Imakita N, Nakano A, Fujita Y, Shichino S, Nakano R, Ueha S, Kasahara K, Muro S, Yano H, Matsushima K, Ito T. Transition of Antibody Titers after SARS-CoV-2 mRNA Vaccination in Japanese Healthcare Workers. Jpn J Infect Dis 2023; 76:72-76. [PMID: 36047181 DOI: 10.7883/yoken.jjid.2022.041] [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: 01/28/2023]
Abstract
Since February 2021, healthcare workers in Japan have been preferentially vaccinated with a messenger RNA vaccine (BNT162b2; Pfizer/BioNTech) against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While many studies have confirmed that this vaccine is highly effective in reducing hospitalization and deaths from coronavirus disease 2019 (COVID-19), antibody titers tend to decline at 3 months after vaccination, leading to a risk of breakthrough infections. Thus, information is needed to support the decision regarding the 3rd vaccination. In this study, we investigated the transition of anti-SARS-CoV-2 spike protein receptor-binding domain (RBD) IgG and neutralizing antibody titers in 37 vaccinated Japanese healthcare workers. Samples were collected 6 times starting before vaccination until 6 months after the second vaccination. The levels of anti-SARS-CoV-2 RBD IgG peaked 1 week after the 2nd vaccination, then declined over time and decreased to < 10% at 6 months after the 2nd vaccination. Additionally, approximately one-third of the healthcare workers were seronegative for the Omicron variant 6 months after the 2nd vaccination. Workers with low anti-SARS-CoV-2 RBD IgG levels also had low neutralizing antibody titers. These data support booster dose administration for healthcare workers, especially those with low anti-SARS-CoV-2 RBD IgG levels.
Collapse
Affiliation(s)
| | | | - Shota Sonobe
- Department of Immunology, Nara Medical University, Japan.,Department of Anesthesiology, Nara Medical University, Japan
| | - Ryutaro Furukawa
- Department of Immunology, Nara Medical University, Japan.,Center for Infectious Diseases, Nara Medical University, Japan
| | - Makiko Konda
- Department of Immunology, Nara Medical University, Japan.,Department of Anesthesiology, Nara Medical University, Japan
| | - Atsushi Hara
- Department of Immunology, Nara Medical University, Japan
| | - Hiroyasu Aoki
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Japan
| | - Yuki Suzuki
- Department of Microbiology and Infectious Diseases, Nara Medical University, Japan
| | - Natsuko Imakita
- Center for Infectious Diseases, Nara Medical University, Japan
| | - Akiyo Nakano
- Department of Microbiology and Infectious Diseases, Nara Medical University, Japan
| | - Yukio Fujita
- Department of Respiratory Medicine, Nara Medical University, Japan
| | - Shigeyuki Shichino
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Japan
| | - Ryuichi Nakano
- Department of Microbiology and Infectious Diseases, Nara Medical University, Japan
| | - Satoshi Ueha
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Japan
| | - Kei Kasahara
- Center for Infectious Diseases, Nara Medical University, Japan
| | - Shigeo Muro
- Department of Respiratory Medicine, Nara Medical University, Japan
| | - Hisakazu Yano
- Department of Microbiology and Infectious Diseases, Nara Medical University, Japan
| | - Kouji Matsushima
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Japan
| | - Toshihiro Ito
- Department of Immunology, Nara Medical University, Japan
| |
Collapse
|
3
|
Iwabuchi S, Tsukahara T, Okayama T, Kitabatake M, Motobayashi H, Shichino S, Imafuku T, Yamaji K, Miyamoto K, Tamura S, Ueha S, Ito T, Murata SI, Kondo T, Ikeo K, Suzuki Y, Matsushima K, Kohara M, Torigoe T, Yamaue H, Hashimoto S. B cell receptor repertoire analysis from autopsy samples of COVID-19 patients. Front Immunol 2023; 14:1034978. [PMID: 36911681 PMCID: PMC9996338 DOI: 10.3389/fimmu.2023.1034978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 02/10/2023] [Indexed: 02/25/2023] Open
Abstract
Neutralizing antibodies against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are being developed world over. We investigated the possibility of producing artificial antibodies from the formalin fixation and paraffin-embedding (FFPE) lung lobes of a patient who died by coronavirus disease 2019 (COVID-19). The B-cell receptors repertoire in the lung tissue where SARS-CoV-2 was detected were considered to have highly sensitive virus-neutralizing activity, and artificial antibodies were produced by combining the most frequently detected heavy and light chains. Some neutralizing effects against the SARS-CoV-2 were observed, and mixing two different artificial antibodies had a higher tendency to suppress the virus. The neutralizing effects were similar to the immunoglobulin G obtained from healthy donors who had received a COVID-19 mRNA vaccine. Therefore, the use of FFPE lung tissue, which preserves the condition of direct virus sensitization, to generate artificial antibodies may be useful against future unknown infectious diseases.
Collapse
Affiliation(s)
- Sadahiro Iwabuchi
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Tomohide Tsukahara
- Department of Pathology, Sapporo Medical University School of Medicine, Hokkaido, Japan
| | - Toshitugu Okayama
- Laboratory of DNA Data Analysis, National Institute of Genetics, Shizuoka, Japan
| | | | - Hideki Motobayashi
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
| | - Shigeyuki Shichino
- Division of Molecular Regulation of Inflammatory and Immune Disease, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Tadashi Imafuku
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Kenzaburo Yamaji
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kyohei Miyamoto
- Department of Emergency and Critical Care Medicine, Wakayama Medical University, Wakayama, Japan
| | - Shinobu Tamura
- Department of Emergency and Critical Care Medicine, Wakayama Medical University, Wakayama, Japan
| | - Satoshi Ueha
- Division of Molecular Regulation of Inflammatory and Immune Disease, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Toshihiro Ito
- Department of Immunology, Nara Medical University, Nara, Japan
| | - Shin-Ichi Murata
- Departments of Human Pathology, Wakayama Medical University, Wakayama, Japan
| | - Toshikazu Kondo
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Kazuho Ikeo
- Laboratory of DNA Data Analysis, National Institute of Genetics, Shizuoka, Japan
| | - Yutaka Suzuki
- Department of Computational Biology, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Kouji Matsushima
- Division of Molecular Regulation of Inflammatory and Immune Disease, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Michinori Kohara
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Toshihiko Torigoe
- Department of Pathology, Sapporo Medical University School of Medicine, Hokkaido, Japan
| | - Hiroki Yamaue
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan.,Departments of Cancer Immunology, Wakayama Medical University, Wakayama, Japan
| | - Shinichi Hashimoto
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
4
|
Ishigaki H, Yasui F, Nakayama M, Endo A, Yamamoto N, Yamaji K, Nguyen CT, Kitagawa Y, Sanada T, Honda T, Munakata T, Higa M, Toyama S, Kono R, Takagi A, Matsumoto Y, Koseki A, Hayashi K, Shiohara M, Ishii K, Saeki Y, Itoh Y, Kohara M. An attenuated vaccinia vaccine encoding the severe acute respiratory syndrome coronavirus-2 spike protein elicits broad and durable immune responses, and protects cynomolgus macaques and human angiotensin-converting enzyme 2 transgenic mice from severe acute respiratory syndrome coronavirus-2 and its variants. Front Microbiol 2022; 13:967019. [PMID: 36466631 PMCID: PMC9716133 DOI: 10.3389/fmicb.2022.967019] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 10/14/2022] [Indexed: 08/27/2023] Open
Abstract
As long as the coronavirus disease-2019 (COVID-19) pandemic continues, new variants of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) with altered antigenicity will emerge. The development of vaccines that elicit robust, broad, and durable protection against SARS-CoV-2 variants is urgently required. We have developed a vaccine consisting of the attenuated vaccinia virus Dairen-I (DIs) strain platform carrying the SARS-CoV-2 S gene (rDIs-S). rDIs-S induced neutralizing antibody and T-lymphocyte responses in cynomolgus macaques and human angiotensin-converting enzyme 2 (hACE2) transgenic mice, and the mouse model showed broad protection against SARS-CoV-2 isolates ranging from the early-pandemic strain (WK-521) to the recent Omicron BA.1 variant (TY38-873). Using a tandem mass tag (TMT)-based quantitative proteomic analysis of lung homogenates from hACE2 transgenic mice, we found that, among mice subjected to challenge infection with WK-521, vaccination with rDIs-S prevented protein expression related to the severe pathogenic effects of SARS-CoV-2 infection (tissue destruction, inflammation, coagulation, fibrosis, and angiogenesis) and restored protein expression related to immune responses (antigen presentation and cellular response to stress). Furthermore, long-term studies in mice showed that vaccination with rDIs-S maintains S protein-specific antibody titers for at least 6 months after a first vaccination. Thus, rDIs-S appears to provide broad and durable protective immunity against SARS-CoV-2, including current variants such as Omicron BA.1 and possibly future variants.
Collapse
Affiliation(s)
- Hirohito Ishigaki
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Fumihiko Yasui
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Misako Nakayama
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Akinori Endo
- Protein Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Naoki Yamamoto
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kenzaburo Yamaji
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Cong Thanh Nguyen
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Yoshinori Kitagawa
- Division of Microbiology and Infectious Diseases, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Takahiro Sanada
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Tomoko Honda
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Tsubasa Munakata
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masahiko Higa
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Sakiko Toyama
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Risa Kono
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Asako Takagi
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yusuke Matsumoto
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Aya Koseki
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kaori Hayashi
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Masanori Shiohara
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Koji Ishii
- Department of Quality Assurance and Radiological Protection, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yasushi Saeki
- Protein Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yasushi Itoh
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
5
|
Sakamoto A, Osawa H, Hashimoto H, Mizuno T, Hasyim AA, Abe YI, Okahashi Y, Ogawa R, Iyori M, Shida H, Yoshida S. A replication-competent smallpox vaccine LC16m8Δ-based COVID-19 vaccine. Emerg Microbes Infect 2022; 11:2359-2370. [PMID: 36069348 PMCID: PMC9527789 DOI: 10.1080/22221751.2022.2122580] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Viral vectors are a potent vaccine platform for inducing humoral and T-cell immune responses. Among the various viral vectors, replication-competent ones are less commonly used for coronavirus disease 2019 (COVID-19) vaccine development compared with replication-deficient ones. Here, we show the availability of a smallpox vaccine LC16m8Δ (m8Δ) as a replication-competent viral vector for a COVID-19 vaccine. M8Δ is a genetically stable variant of the licensed and highly effective Japanese smallpox vaccine LC16m8. Here, we generated two m8Δ recombinants: one harbouring a gene cassette encoding the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) glycoprotein, named m8Δ-SARS2(P7.5-S)-HA; and one encoding the S protein with a highly polybasic motif at the S1/S2 cleavage site, named m8Δ-SARS2(P7.5-SHN)-HA. M8Δ-SARS2(P7.5-S)-HA induced S-specific antibodies in mice that persisted for at least six weeks after a homologous boost immunization. All eight analysed serum samples displayed neutralizing activity against an S-pseudotyped virus at a level similar to that of serum samples from patients with COVID-19, and more than half (5/8) also had neutralizing activity against the Delta/B.1.617.2 variant of concern. Importantly, most serum samples also neutralized the infectious SARS-CoV-2 Wuhan and Delta/B.1.617.2 strains. In contrast, immunization with m8Δ-SARS2(P7.5-SHN)-HA elicited significantly lower antibody titres, and the induced antibodies had less neutralizing activity. Regarding T-cell immunity, both m8Δ recombinants elicited S-specific multifunctional CD8+ and CD4+ T-cell responses even after just a primary immunization. Thus, m8Δ provides an alternative method for developing a novel COVID-19 vaccine.
Collapse
Affiliation(s)
- Akihiko Sakamoto
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Ishikawa, Japan
| | - Hiroaki Osawa
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Ishikawa, Japan
| | - Hinata Hashimoto
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Ishikawa, Japan
| | - Tetsushi Mizuno
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Ishikawa, Japan.,Department of Global Infectious Diseases, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Ammar A Hasyim
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Ishikawa, Japan
| | - Yu-Ichi Abe
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Ishikawa, Japan
| | - Yuto Okahashi
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Ishikawa, Japan
| | - Ryohei Ogawa
- Department of Radiology, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Mitsuhiro Iyori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Ishikawa, Japan
| | - Hisatoshi Shida
- Division of Molecular Virology, Institute of Immunological Science, Hokkaido University, Sapporo, Japan
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Ishikawa, Japan
| |
Collapse
|
6
|
dos-Santos JS, Firmino-Cruz L, da Fonseca-Martins AM, Oliveira-Maciel D, Perez GG, Roncaglia-Pereira VA, Dumard CH, Guedes-da-Silva FH, Santos ACV, Leandro MDS, Ferreira JRM, Guimarães-Pinto K, Conde L, Rodrigues DAS, Silva MVDM, Alvim RGF, Lima TM, Marsili FF, Abreu DPB, Ferreira Jr. OC, Mohana Borges RDS, Tanuri A, Souza TML, Rossi-Bergmann B, Vale AM, Silva JL, de Oliveira AC, Filardy AD, Gomes AMO, de Matos Guedes HL. Immunogenicity of SARS-CoV-2 Trimeric Spike Protein Associated to Poly(I:C) Plus Alum. Front Immunol 2022; 13:884760. [PMID: 35844561 PMCID: PMC9281395 DOI: 10.3389/fimmu.2022.884760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/15/2022] [Indexed: 12/20/2022] Open
Abstract
The SARS-CoV-2 pandemic has had a social and economic impact worldwide, and vaccination is an efficient strategy for diminishing those damages. New adjuvant formulations are required for the high vaccine demands, especially adjuvant formulations that induce a Th1 phenotype. Herein we assess a vaccination strategy using a combination of Alum and polyinosinic:polycytidylic acid [Poly(I:C)] adjuvants plus the SARS-CoV-2 spike protein in a prefusion trimeric conformation by an intradermal (ID) route. We found high levels of IgG anti-spike antibodies in the serum by enzyme linked immunosorbent assay (ELISA) and high neutralizing titers against SARS-CoV-2 in vitro by neutralization assay, after two or three immunizations. By evaluating the production of IgG subtypes, as expected, we found that formulations containing Poly(I:C) induced IgG2a whereas Alum did not. The combination of these two adjuvants induced high levels of both IgG1 and IgG2a. In addition, cellular immune responses of CD4+ and CD8+ T cells producing interferon-gamma were equivalent, demonstrating that the Alum + Poly(I:C) combination supported a Th1 profile. Based on the high neutralizing titers, we evaluated B cells in the germinal centers, which are specific for receptor-binding domain (RBD) and spike, and observed that more positive B cells were induced upon the Alum + Poly(I:C) combination. Moreover, these B cells produced antibodies against both RBD and non-RBD sites. We also studied the impact of this vaccination preparation [spike protein with Alum + Poly(I:C)] in the lungs of mice challenged with inactivated SARS-CoV-2 virus. We found a production of IgG, but not IgA, and a reduction in neutrophil recruitment in the bronchoalveolar lavage fluid (BALF) of mice, suggesting that our immunization scheme reduced lung inflammation. Altogether, our data suggest that Alum and Poly(I:C) together is a possible adjuvant combination for vaccines against SARS-CoV-2 by the intradermal route.
Collapse
Affiliation(s)
- Júlio Souza dos-Santos
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Luan Firmino-Cruz
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Alessandra Marcia da Fonseca-Martins
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Diogo Oliveira-Maciel
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Gustavo Guadagnini Perez
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Victor A. Roncaglia-Pereira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Carlos H. Dumard
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Francisca H. Guedes-da-Silva
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Ana C. Vicente Santos
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Monique dos Santos Leandro
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | | | - Kamila Guimarães-Pinto
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Luciana Conde
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Danielle A. S. Rodrigues
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | | | - Renata G. F. Alvim
- Cell Culture Engineering Lab., Alberto Luiz Coimbra Institute for Graduate Studies and Research in Engineering (COPPE), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Tulio M. Lima
- Cell Culture Engineering Lab., Alberto Luiz Coimbra Institute for Graduate Studies and Research in Engineering (COPPE), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Federico F. Marsili
- Cell Culture Engineering Lab., Alberto Luiz Coimbra Institute for Graduate Studies and Research in Engineering (COPPE), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Daniel P. B. Abreu
- Cell Culture Engineering Lab., Alberto Luiz Coimbra Institute for Graduate Studies and Research in Engineering (COPPE), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | | | | | - Amilcar Tanuri
- National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Institute of Biology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Thiago Moreno L. Souza
- Immunopharmacology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
- National Institute for Science and Technology on Innovation in Diseases of Neglected Populations (INCT/IDPN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Bartira Rossi-Bergmann
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - André M. Vale
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Jerson Lima Silva
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Andréa Cheble de Oliveira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | | | - Andre M. O. Gomes
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Herbert Leonel de Matos Guedes
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Interdisciplinary Medical Research Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| |
Collapse
|
7
|
Harbour JC, Lyski ZL, Schell JB, Thomas A, Messer WB, Slifka MK, Nolz JC. Cellular and Humoral Immune Responses in Mice Immunized with Vaccinia Virus Expressing the SARS-CoV-2 Spike Protein. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:2596-2604. [PMID: 33972374 PMCID: PMC8165000 DOI: 10.4049/jimmunol.2100054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/22/2021] [Indexed: 12/26/2022]
Abstract
The COVID-19 pandemic is a global health emergency, and the development of a successful vaccine will ultimately be required to prevent the continued spread and seasonal recurrence of this disease within the human population. However, very little is known about either the quality of the adaptive immune response or the viral Ag targets that will be necessary to prevent the spread of the infection. In this study, we generated recombinant Vaccinia virus expressing the full-length spike protein from SARS-CoV-2 (VacV-S) to evaluate the cellular and humoral immune response mounted against this viral Ag in mice. Both CD8+ and CD4+ T cells specific to the SARS-CoV-2 spike protein underwent robust expansion, contraction, and persisted for at least 40 d following a single immunization with VacV-S. Vaccination also caused the rapid emergence of spike-specific IgG-neutralizing Abs. Interestingly, both the cellular and humoral immune responses strongly targeted the S1 domain of spike following VacV-S immunization. Notably, immunization with VacV-expressing spike conjugated to the MHC class II invariant chain, a strategy previously reported by us and others to enhance the immunogenicity of antigenic peptides, did not promote stronger spike-specific T cell or Ab responses in vivo. Overall, these findings demonstrate that an immunization approach using VacV or attenuated versions of VacV expressing the native, full-length SARS-CoV-2 spike protein could be used for further vaccine development to prevent the spread of COVID-19.
Collapse
Affiliation(s)
- Jake C Harbour
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR
| | - Zoe L Lyski
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR
| | - John B Schell
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR
| | - Archana Thomas
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - William B Messer
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR
- Department of Medicine, Division of Infectious Diseases, Oregon Health & Science University, Portland, OR
- Program in Epidemiology, Oregon Health & Science University-Portland State University School of Public Health, Oregon Health & Science University, Portland, OR
| | - Mark K Slifka
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Jeffrey C Nolz
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR;
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR; and
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR
| |
Collapse
|
8
|
Lima I, Cino EA. Sequence similarity in 3D for comparison of protein families. J Mol Graph Model 2021; 106:107906. [PMID: 33848948 DOI: 10.1016/j.jmgm.2021.107906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/18/2021] [Accepted: 03/18/2021] [Indexed: 11/26/2022]
Abstract
Homologous proteins are often compared by pairwise sequence alignment, and structure superposition if the atomic coordinates are available. Unification of sequence and structure data is an important task in structural biology. Here, we present the Sequence Similarity 3D (SS3D) method of integrating sequence and structure information. SS3D is a distance and substitution matrix-based method for straightforward visualization of regions of similarity and difference between homologous proteins. This work details the SS3D approach, and demonstrates its utility through case studies comparing members of several protein families. The examples show that SS3D can effectively highlight biologically important regions of similarity and dissimilarity. We anticipate that the method will be useful for numerous structural biology applications, including, but not limited to, studies of binding specificity, structure-function relationships, and evolutionary pathways. SS3D is available with a manual and tutorial at https://github.com/0x462e41/SS3D/.
Collapse
Affiliation(s)
- Igor Lima
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Elio A Cino
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Brazil.
| |
Collapse
|
9
|
Sensitization with vaccinia virus encoding H5N1 hemagglutinin restores immune potential against H5N1 influenza virus. Sci Rep 2016; 6:37915. [PMID: 27892498 PMCID: PMC5124960 DOI: 10.1038/srep37915] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 11/04/2016] [Indexed: 11/08/2022] Open
Abstract
H5N1 highly pathogenic avian influenza (H5N1 HPAI) virus causes elevated mortality compared with seasonal influenza viruses like H1N1 pandemic influenza (H1N1 pdm) virus. We identified a mechanism associated with the severe symptoms seen with H5N1 HPAI virus infection. H5N1 HPAI virus infection induced a decrease of dendritic cell number in the splenic extrafollicular T-cell zone and impaired formation of the outer layers of B-cell follicles, resulting in insufficient levels of antibody production after infection. However, in animals vaccinated with a live recombinant vaccinia virus expressing the H5 hemagglutinin, infection with H5N1 HPAI virus induced parafollicular dendritic cell accumulation and efficient antibody production. These results indicate that a recombinant vaccinia encoding H5 hemagglutinin gene does not impair dendritic cell recruitment and can be a useful vaccine candidate.
Collapse
|
10
|
Sánchez-Sampedro L, Perdiguero B, Mejías-Pérez E, García-Arriaza J, Di Pilato M, Esteban M. The evolution of poxvirus vaccines. Viruses 2015; 7:1726-803. [PMID: 25853483 PMCID: PMC4411676 DOI: 10.3390/v7041726] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/16/2015] [Accepted: 03/27/2015] [Indexed: 02/07/2023] Open
Abstract
After Edward Jenner established human vaccination over 200 years ago, attenuated poxviruses became key players to contain the deadliest virus of its own family: Variola virus (VARV), the causative agent of smallpox. Cowpox virus (CPXV) and horsepox virus (HSPV) were extensively used to this end, passaged in cattle and humans until the appearance of vaccinia virus (VACV), which was used in the final campaigns aimed to eradicate the disease, an endeavor that was accomplished by the World Health Organization (WHO) in 1980. Ever since, naturally evolved strains used for vaccination were introduced into research laboratories where VACV and other poxviruses with improved safety profiles were generated. Recombinant DNA technology along with the DNA genome features of this virus family allowed the generation of vaccines against heterologous diseases, and the specific insertion and deletion of poxvirus genes generated an even broader spectrum of modified viruses with new properties that increase their immunogenicity and safety profile as vaccine vectors. In this review, we highlight the evolution of poxvirus vaccines, from first generation to the current status, pointing out how different vaccines have emerged and approaches that are being followed up in the development of more rational vaccines against a wide range of diseases.
Collapse
MESH Headings
- Animals
- History, 18th Century
- History, 19th Century
- History, 20th Century
- History, 21st Century
- Humans
- Poxviridae/immunology
- Poxviridae/isolation & purification
- Smallpox/prevention & control
- Smallpox Vaccine/history
- Smallpox Vaccine/immunology
- Smallpox Vaccine/isolation & purification
- Vaccines, Attenuated/history
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/isolation & purification
- Vaccines, Synthetic/history
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/isolation & purification
Collapse
Affiliation(s)
- Lucas Sánchez-Sampedro
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| | - Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| | - Ernesto Mejías-Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain
| | - Mauro Di Pilato
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid-28049, Spain.
| |
Collapse
|
11
|
Kidokoro M, Shida H. Vaccinia Virus LC16m8∆ as a Vaccine Vector for Clinical Applications. Vaccines (Basel) 2014; 2:755-71. [PMID: 26344890 PMCID: PMC4494248 DOI: 10.3390/vaccines2040755] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 09/16/2014] [Accepted: 09/28/2014] [Indexed: 01/14/2023] Open
Abstract
The LC16m8 strain of vaccinia virus, the active ingredient in the Japanese smallpox vaccine, was derived from the Lister/Elstree strain. LC16m8 is replication-competent and has been administered to over 100,000 infants and 3,000 adults with no serious adverse reactions. Despite this outstanding safety profile, the occurrence of spontaneously-generated large plaque-forming virulent LC16m8 revertants following passage in cell culture is a major drawback. We identified the gene responsible for the reversion and deleted the gene (B5R) from LC16m8 to derive LC16m8Δ. LC16m8∆ is non-pathogenic in immunodeficient severe combined immunodeficiency (SCID) mice, genetically-stable and does not reverse to a large-plaque phenotype upon passage in cell culture, even under conditions in which most LC16m8 populations are replaced by revertants. Moreover, LC16m8∆ is >500-fold more effective than the non-replicating vaccinia virus (VV), Modified Vaccinia Ankara (MVA), at inducing murine immune responses against pathogenic VV. LC16m8∆, which expresses the SIV gag gene, also induced anti-Gag CD8⁺ T-cells more efficiently than MVA and another non-replicating VV, Dairen I minute-pock variants (DIs). Moreover, LC16m8∆ expressing HIV-1 Env in combination with a Sendai virus vector induced the production of anti-Env antibodies and CD8⁺ T-cells. Thus, the safety and efficacy of LC16m8∆ mean that it represents an outstanding platform for the development of human vaccine vectors.
Collapse
Affiliation(s)
- Minoru Kidokoro
- Department of Virology III, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama-shi, Tokyo 208-0011, Japan.
| | - Hisatoshi Shida
- Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo 060-0815, Japan.
| |
Collapse
|
12
|
Kwon JS, Yoon J, Kim YJ, Kang K, Woo S, Jung DI, Song MK, Kim EH, Kwon HI, Choi YK, Kim J, Lee J, Yoon Y, Shin EC, Youn JW. Vaccinia-based influenza vaccine overcomes previously induced immunodominance hierarchy for heterosubtypic protection. Eur J Immunol 2014; 44:2360-9. [PMID: 24825439 DOI: 10.1002/eji.201344005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Revised: 03/21/2014] [Accepted: 05/07/2014] [Indexed: 12/27/2022]
Abstract
Growing concerns about unpredictable influenza pandemics require a broadly protective vaccine against diverse influenza strains. One of the promising approaches was a T cell-based vaccine, but the narrow breadth of T-cell immunity due to the immunodominance hierarchy established by previous influenza infection and efficacy against only mild challenge condition are important hurdles to overcome. To model T-cell immunodominance hierarchy in humans in an experimental setting, influenza-primed C57BL/6 mice were chosen and boosted with a mixture of vaccinia recombinants, individually expressing consensus sequences from avian, swine, and human isolates of influenza internal proteins. As determined by IFN-γ ELISPOT and polyfunctional cytokine secretion, the vaccinia recombinants of influenza expanded the breadth of T-cell responses to include subdominant and even minor epitopes. Vaccine groups were successfully protected against 100 LD50 challenges with PR/8/34 and highly pathogenic avian influenza H5N1, which contained the identical dominant NP366 epitope. Interestingly, in challenge with pandemic A/Cal/04/2009 containing mutations in the dominant epitope, only the group vaccinated with rVV-NP + PA showed improved protection. Taken together, a vaccinia-based influenza vaccine expressing conserved internal proteins improved the breadth of influenza-specific T-cell immunity and provided heterosubtypic protection against immunologically close as well as distant influenza strains.
Collapse
Affiliation(s)
- Ji-Sun Kwon
- Vaccine II, Mogam Biotechnology Research Institute, Yongin Si, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Abstract
Middle East respiratory syndrome (MERS) is a newly emerging infectious disease caused by a novel coronavirus, MERS-coronavirus (MERS-CoV), a new member in the lineage C of β-coronavirus (β-CoV). The increased human cases and high mortality rate of MERS-CoV infection make it essential to develop safe and effective vaccines. In this review, the current advancements and potential strategies in the development of MERS vaccines, particularly subunit vaccines based on MERS-CoV spike (S) protein and its receptor-binding domain (RBD), are discussed. How to improve the efficacy of subunit vaccines through novel adjuvant formulations and routes of administration as well as currently available animal models for evaluating the in vivo efficacy of MERS-CoV vaccines are also addressed. Overall, these strategies may have important implications for the development of effective and safe vaccines for MERS-CoV in the future.
Collapse
Affiliation(s)
- Naru Zhang
- Lindsley F. Kimball Research Institute, New York Blood Center,New York, NY,USA
| | - Shibo Jiang
- Lindsley F. Kimball Research Institute, New York Blood Center,New York, NY,USA
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University,Shanghai,China
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center,New York, NY,USA
| |
Collapse
|
14
|
Caufour P, Rufael T, Lamien CE, Lancelot R, Kidane M, Awel D, Sertse T, Kwiatek O, Libeau G, Sahle M, Diallo A, Albina E. Protective efficacy of a single immunization with capripoxvirus-vectored recombinant peste des petits ruminants vaccines in presence of pre-existing immunity. Vaccine 2014; 32:3772-9. [PMID: 24837763 DOI: 10.1016/j.vaccine.2014.05.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 04/25/2014] [Accepted: 05/01/2014] [Indexed: 11/30/2022]
Abstract
Sheeppox, goatpox and peste des petits ruminants (PPR) are highly contagious ruminant diseases widely distributed in Africa, the Middle East and Asia. Capripoxvirus (CPV)-vectored recombinant PPR vaccines (rCPV-PPR vaccines), which have been developed and shown to protect against both Capripox (CP) and PPR, would be critical tools in the control of these important diseases. In most parts of the world, these disease distributions overlap each other leaving concerns about the potential impact that pre-existing immunity against either disease may have on the protective efficacy of these bivalent rCPV-PPR vaccines. Currently, this question has not been indisputably addressed. Therefore, we undertook this study, under experimental conditions designed for the context of mass vaccination campaigns of small ruminants, using the two CPV recombinants (Kenya sheep-1 (KS-1) strain-based constructs) developed previously in our laboratory. Pre-existing immunity was first induced by immunization either with an attenuated CPV vaccine strain (KS-1) or the attenuated PPRV vaccine strain (Nigeria 75/1) and animals were thereafter inoculated once subcutaneously with a mixture of CPV recombinants expressing either the hemagglutinin (H) or the fusion (F) protein gene of PPRV (10(3) TCID50/animal of each). Finally, these animals were challenged with a virulent CPV strain followed by a virulent PPRV strain 3 weeks later. Our study demonstrated full protection against CP for vaccinated animals with prior exposure to PPRV and a partial protection against PPR for vaccinated animals with prior exposure to CPV. The latter animals exhibited a mild clinical form of PPR and did not show any post-challenge anamnestic neutralizing antibody response against PPRV. The implications of these results are discussed herein and suggestions made for future research regarding the development of CPV-vectored vaccines.
Collapse
Affiliation(s)
- Philippe Caufour
- CIRAD, UMR CMAEE, F-34398 Montpellier, France; INRA, UMR1309 CMAEE, F-34398 Montpellier, France.
| | - Tesfaye Rufael
- National Animal health Diagnosis and Investigation Center (NAHDIC), P.O. Box 04, Sebeta, Ethiopia
| | - Charles Euloge Lamien
- Animal Production and Health Laboratory, FAO/IAEA Agriculture & Biotechnology Laboratory, IAEA Laboratories, International Atomic Energy Agency, Wagramer Strasse 5, P.O. Box 100, A-1400 Vienna, Austria
| | - Renaud Lancelot
- CIRAD, UMR CMAEE, F-34398 Montpellier, France; INRA, UMR1309 CMAEE, F-34398 Montpellier, France
| | - Menbere Kidane
- National Animal health Diagnosis and Investigation Center (NAHDIC), P.O. Box 04, Sebeta, Ethiopia
| | - Dino Awel
- National Animal health Diagnosis and Investigation Center (NAHDIC), P.O. Box 04, Sebeta, Ethiopia
| | - Tefera Sertse
- National Animal health Diagnosis and Investigation Center (NAHDIC), P.O. Box 04, Sebeta, Ethiopia
| | - Olivier Kwiatek
- CIRAD, UMR CMAEE, F-34398 Montpellier, France; INRA, UMR1309 CMAEE, F-34398 Montpellier, France
| | - Geneviève Libeau
- CIRAD, UMR CMAEE, F-34398 Montpellier, France; INRA, UMR1309 CMAEE, F-34398 Montpellier, France
| | - Mesfin Sahle
- National Animal health Diagnosis and Investigation Center (NAHDIC), P.O. Box 04, Sebeta, Ethiopia
| | - Adama Diallo
- Animal Production and Health Laboratory, FAO/IAEA Agriculture & Biotechnology Laboratory, IAEA Laboratories, International Atomic Energy Agency, Wagramer Strasse 5, P.O. Box 100, A-1400 Vienna, Austria
| | - Emmanuel Albina
- INRA, UMR1309 CMAEE, F-34398 Montpellier, France; CIRAD, UMR CMAEE, F-97170 Petit-Bourg, Guadeloupe, France
| |
Collapse
|
15
|
Phagocytic cells contribute to the antibody-mediated elimination of pulmonary-infected SARS coronavirus. Virology 2014; 454-455:157-68. [PMID: 24725942 PMCID: PMC7111974 DOI: 10.1016/j.virol.2014.02.005] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 01/10/2014] [Accepted: 02/04/2014] [Indexed: 12/28/2022]
Abstract
While the 2002–2003 outbreak of severe acute respiratory syndrome (SARS) resulted in 774 deaths, patients who were affected with mild pulmonary symptoms successfully recovered. The objective of the present work was to identify, using SARS coronavirus (SARS-CoV) mouse infection models, immune factors responsible for clearing of the virus. The elimination of pulmonary SARS-CoV infection required the activation of B cells by CD4+ T cells. Furthermore, passive immunization (post-infection) with homologous (murine) anti-SARS-CoV antiserum showed greater elimination efficacy against SARS-CoV than that with heterologous (rabbit) antiserum, despite the use of equivalent titers of neutralizing antibodies. This distinction was mediated by mouse phagocytic cells (monocyte-derived infiltrating macrophages and partially alveolar macrophages, but not neutrophils), as demonstrated both by adoptive transfer from donors and by immunological depletion of selected cell types. These results indicate that the cooperation of anti-SARS-CoV antibodies and phagocytic cells plays an important role in the elimination of SARS-CoV.
Collapse
|
16
|
Immunization with a recombinant vaccinia virus that encodes nonstructural proteins of the hepatitis C virus suppresses viral protein levels in mouse liver. PLoS One 2012; 7:e51656. [PMID: 23284733 PMCID: PMC3524174 DOI: 10.1371/journal.pone.0051656] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 11/05/2012] [Indexed: 12/16/2022] Open
Abstract
Chronic hepatitis C, which is caused by infection with the hepatitis C virus (HCV), is a global health problem. Using a mouse model of hepatitis C, we examined the therapeutic effects of a recombinant vaccinia virus (rVV) that encodes an HCV protein. We generated immunocompetent mice that each expressed multiple HCV proteins via a Cre/loxP switching system and established several distinct attenuated rVV strains. The HCV core protein was expressed consistently in the liver after polyinosinic acid–polycytidylic acid injection, and these mice showed chronic hepatitis C-related pathological findings (hepatocyte abnormalities, accumulation of glycogen, steatosis), liver fibrosis, and hepatocellular carcinoma. Immunization with one rVV strain (rVV-N25), which encoded nonstructural HCV proteins, suppressed serum inflammatory cytokine levels and alleviated the symptoms of pathological chronic hepatitis C within 7 days after injection. Furthermore, HCV protein levels in liver tissue also decreased in a CD4 and CD8 T-cell-dependent manner. Consistent with these results, we showed that rVV-N25 immunization induced a robust CD8 T-cell immune response that was specific to the HCV nonstructural protein 2. We also demonstrated that the onset of chronic hepatitis in CN2-29(+/−)/MxCre(+/−) mice was mainly attributable to inflammatory cytokines, (tumor necrosis factor) TNF-α and (interleukin) IL-6. Thus, our generated mice model should be useful for further investigation of the immunological processes associated with persistent expression of HCV proteins because these mice had not developed immune tolerance to the HCV antigen. In addition, we propose that rVV-N25 could be developed as an effective therapeutic vaccine.
Collapse
|
17
|
Chen YZ, Liu G, Senju S, Wang Q, Irie A, Haruta M, Matsui M, Yasui F, Kohara M, Nishimura Y. Identification of SARS-COV spike protein-derived and HLA-A2-restricted human CTL epitopes by using a new muramyl dipeptidederivative adjuvant. Int J Immunopathol Pharmacol 2010; 23:165-77. [PMID: 20378004 DOI: 10.1177/039463201002300115] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Severe acute respiratory syndrome (SARS) spread during the winter of 2003, and attempts have been made to develop vaccines against SARS corona virus (SARS-CoV). The present study provides a strategy to rapidly identify SARS-CoV-derived antigenic peptides recognized by HLA-A2-restricted cytotoxic T lymphocytes (CTLs). Forty-three candidate peptides having HLA-A2-binding motifs were selected in silico and HLA-A2/Db chimeric MHC class I-transgenic mice were immunized with these peptides and a new derivative of muramyl dipeptide that can induce upregulation of HLA-DR, CD80, CD86, and CD40 in human CD14+ antigen presenting cells, was administered as an adjuvant. Six HLAA2-restricted mouse CTL epitopes were identified, including two new epitopes which have never been reported before. One of the novel peptides was naturally processed and successfully induced HLAA2-restricted specific CTLs in both HLA transgenic mice and healthy donors. The method was useful, convenient and efficient for rapid identification of CTL epitopes derived from SARS-CoV proteins and will be possibly applicable for other pathogens to develop a peptide-based vaccine.
Collapse
Affiliation(s)
- Y-Z Chen
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Yasui F, Kai C, Saito K, Inoue S, Yoneda M, Morita K, Mizuno K, Kohara M. Analysis of the mechanism by which BALB/c mice having prior immunization with nucleocapsid protein of SARS-CoV develop severe pneumonia after SARS-CoV infection. ACTA ACUST UNITED AC 2010; 2:44-50. [PMID: 32288911 PMCID: PMC7128161 DOI: 10.1016/j.provac.2010.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The precise mechanism of severe acute respiratory syndrome (SARS), which is caused by SARS-associated coronavirus (SARS-CoV), is still unclear. We generated recombinant vaccinia virus (rVV) LC16m8 strain which simultaneously expresses four structural proteins of SARS-CoV, including nucleocapsid (N), membrane (M), envelop (E), spike (S) proteins (rVV-NMES) and reported that old BALB/c mice having prior immunization with rVV-NMES develop severe pneumonia similar to those of control mice though rVV-NMES-immunized mice showed lower pulmonary viral titer than in the control mice. Furthermore, we determined which SARS-CoV structural protein for the prior rVV-immunization is responsible for the severe pneumonia after the SARS-CoV infection as observed in the rVV-NMES-immunized mice. Old BALB/c mice were inoculated intradermally with rVV that expressed each structural proteins of SARS-CoV (rVV-N, -M, -E, or -S) with or without rVV-S and then infected intranasally with SARS-CoV more than 4 weeks later. At 9 days after SARS-CoV infection, the rVV-N-immunized mice show more severe pneumonia than in other groups. Furthermore, significant up-regulation of Th1 (IL-2)- and Th2 (IL-4 and IL-5)-bias cytokines and down-regulation of anti-inflammatory cytokine (IL-10 and TGF-β) were observed in rVV-N-immunized mice, resulting in the intensive infiltration of immunocompetent cells into the lung. In contrast, rVV-S-immunized mice showed only low pulmonary viral tier and slight pneumonia. However, the mice having co-immunization with both rVV-N and rVV-S showed severe pneumonia though their pulmonary viral titer was low. These results suggest that an excessive host immune response against the N protein of SARS-CoV is involved in severe pneumonia caused by SARS-CoV infection. These findings increase our understanding of the pathogenesis of SARS.
Collapse
Affiliation(s)
- Fumihiko Yasui
- Dep. Micobiol. & Cell Biol., Tokyo Metro. Inst. Med. Sci
| | - Chieko Kai
- Lab. Animal Res. Center, The Inst. Med. Sci., Univ. of Tokyo
| | - Kousuke Saito
- Dep. Micobiol. & Cell Biol., Tokyo Metro. Inst. Med. Sci
| | - Shingo Inoue
- Dep. of Virol., Inst. of Tropic. Med., Nagasaki Univ
| | - Misako Yoneda
- Lab. Animal Res. Center, The Inst. Med. Sci., Univ. of Tokyo
| | | | | | | |
Collapse
|
19
|
Poh WP, Narasaraju T, Pereira NA, Zhong F, Phoon MC, Macary PA, Wong SH, Lu J, Koh DR, Chow VTK. Characterization of cytotoxic T-lymphocyte epitopes and immune responses to SARS coronavirus spike DNA vaccine expressing the RGD-integrin-binding motif. J Med Virol 2009; 81:1131-9. [PMID: 19475608 PMCID: PMC7166745 DOI: 10.1002/jmv.21571] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Integrins are critical for initiating T‐cell activation events. The integrin‐binding motif Arg‐Gly‐Asp (RGD) was incorporated into the pcDNA 3.1 mammalian expression vector expressing the codon‐optimized extracellular domain of SARS coronavirus (SARS‐CoV) spike protein, and tested by immunizing C57BL/6 mice. Significant cell‐mediated immune responses were characterized by cytotoxic T‐lymphocyte 51Cr release assay and interferon‐gamma secretion ELISPOT assay against RMA‐S target cells presenting predicted MHC class I H2‐Kb epitopes, including those spanning residues 884–891 and 1116–1123 within the S2 subunit of SARS‐CoV spike protein. DNA vaccines incorporating the Spike‐RGD/His motif or the Spike‐His construct generated robust cell‐mediated immune responses. Moreover, the Spike‐His DNA vaccine construct generated a significant antibody response. Immunization with these DNA vaccine constructs elicited significant cellular and humoral immune responses. Additional T‐cell epitopes within the SARS‐CoV spike protein that may contribute to cell‐mediated immunity in vivo were also identified. J. Med. Virol. 81:1131–1139, 2009. © 2009 Wiley‐Liss, Inc.
Collapse
Affiliation(s)
- W P Poh
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore, Singapore
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Pimentel TAPF, Yan Z, Jeffers SA, Holmes KV, Hodges RS, Burkhard P. Peptide nanoparticles as novel immunogens: design and analysis of a prototypic severe acute respiratory syndrome vaccine. Chem Biol Drug Des 2009; 73:53-61. [PMID: 19152635 PMCID: PMC2756483 DOI: 10.1111/j.1747-0285.2008.00746.x] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Severe acute respiratory syndrome (SARS) is an infectious disease caused by a novel coronavirus that cost nearly 800 lives. While there have been no recent outbreaks of the disease, the threat remains as SARS coronavirus (SARS‐CoV) like strains still exist in animal reservoirs. Therefore, the development of a vaccine against SARS is in grave need. Here, we have designed and produced a prototypic SARS vaccine: a self‐assembling polypeptide nanoparticle that repetitively displays a SARS B‐cell epitope from the C‐terminal heptad repeat of the virus’ spike protein. Biophysical analyses with circular dichroism, transmission electron microscopy and dynamic light scattering confirmed the computational design showing α‐helcial nanoparticles with sizes of about 25 nm. Immunization experiments with no adjuvants were performed with BALB/c mice. An investigation of the binding properties of the elicited antibodies showed that they were highly conformation specific for the coiled‐coil epitope because they specifically recognized the native trimeric conformation of C‐terminal heptad repeat region. Consequently, the antisera exhibited neutralization activity in an in vitro infection inhibition assay. We conclude that these peptide nanoparticles represent a promising platform for vaccine design, in particular for diseases that are characterized by neutralizing epitopes with coiled‐coil conformation such as SARS‐CoV or other enveloped viruses.
Collapse
Affiliation(s)
- Tais A P F Pimentel
- Institute of Materials Science, University of Connecticut, Storrs, CT 06269-3136, USA
| | | | | | | | | | | |
Collapse
|
21
|
Yasui F, Kai C, Kitabatake M, Inoue S, Yoneda M, Yokochi S, Kase R, Sekiguchi S, Morita K, Hishima T, Suzuki H, Karamatsu K, Yasutomi Y, Shida H, Kidokoro M, Mizuno K, Matsushima K, Kohara M. Prior immunization with severe acute respiratory syndrome (SARS)-associated coronavirus (SARS-CoV) nucleocapsid protein causes severe pneumonia in mice infected with SARS-CoV. THE JOURNAL OF IMMUNOLOGY 2009; 181:6337-48. [PMID: 18941225 DOI: 10.4049/jimmunol.181.9.6337] [Citation(s) in RCA: 197] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The details of the mechanism by which severe acute respiratory syndrome-associated coronavirus (SARS-CoV) causes severe pneumonia are unclear. We investigated the immune responses and pathologies of SARS-CoV-infected BALB/c mice that were immunized intradermally with recombinant vaccinia virus (VV) that expressed either the SARS-CoV spike (S) protein (LC16m8rVV-S) or simultaneously all the structural proteins, including the nucleocapsid (N), membrane (M), envelope (E), and S proteins (LC16m8rVV-NMES) 7-8 wk before intranasal SARS-CoV infection. The LC16m8rVV-NMES-immunized group exhibited as severe pneumonia as the control groups, although LC16m8rVV-NMES significantly decreased the pulmonary SARS-CoV titer to the same extent as LC16m8rVV-S. To identify the cause of the exacerbated pneumonia, BALB/c mice were immunized with recombinant VV that expressed the individual structural proteins of SARS-CoV (LC16mOrVV-N, -M, -E, -S) with or without LC16mOrVV-S (i.e., LC16mOrVV-N, LC16mOrVV-M, LC16mOrVV-E, or LC16mOrVV-S alone or LC16mOrVV-N + LC16mOrVV-S, LC16mOrVV-M + LC16mOrVV-S, or LC16mOrVV-E + LC16mOrVV-S), and infected with SARS-CoV more than 4 wk later. Both LC16mOrVV-N-immunized mice and LC16mOrVV-N + LC16mOrVV-S-immunized mice exhibited severe pneumonia. Furthermore, LC16mOrVV-N-immunized mice upon infection exhibited significant up-regulation of both Th1 (IFN-gamma, IL-2) and Th2 (IL-4, IL-5) cytokines and down-regulation of anti-inflammatory cytokines (IL-10, TGF-beta), resulting in robust infiltration of neutrophils, eosinophils, and lymphocytes into the lung, as well as thickening of the alveolar epithelium. These results suggest that an excessive host immune response against the nucleocapsid protein of SARS-CoV is involved in severe pneumonia caused by SARS-CoV infection. These findings increase our understanding of the pathogenesis of SARS.
Collapse
Affiliation(s)
- Fumihiko Yasui
- Department of Microbiology and Cell Biology, The Tokyo Metropolitan Institute of Medical Science, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Current World Literature. Curr Opin Pulm Med 2008; 14:266-73. [DOI: 10.1097/mcp.0b013e3282ff8c19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|