1
|
Mirjalili SZ, Tamiji Z, Shirangi M, Amini M. A GC-MS Method for Determination of β-Propiolactone Residues in Inactivated Covid-19 Vaccines. J Chromatogr Sci 2024; 62:905-911. [PMID: 39286865 DOI: 10.1093/chromsci/bmae049] [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: 03/06/2023] [Revised: 05/23/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024]
Abstract
β-propiolactone is a common inactivator agent used in vaccines. Due to β-propiolactone carcinogenicity, complete hydrolysis of it is necessary to prevent cytotoxicity in mammalian cells. As a result, more attention should be paid to it at the clinic, and it is important to measure its trace amounts. β-propiolactone analysis is challenging due to its instability. A simple and fast gas chromatography-mass spectrometry method was developed for quantitation of residual β-propiolactone in inactivated coronavirus disease 2019 (Covid-19) vaccines. Caprolactone was used as an internal standard in sample solutions; the analysis was performed after extraction of analyte from vaccine media by ethyl acetate. The validity of the method was studied with a linearity of r2 > 0.99 over the concentration range of 0.2-20 μg/mL with the limit of detection and the limit of quantification of 0.07 and 0.20 μg/mL, respectively. The target analyte β-propiolactone was not detected in the samples, demonstrating the test samples were qualified. The established method can be used for quality control of inactivated Covid-19 vaccines.
Collapse
Affiliation(s)
- Seyedeh Zohreh Mirjalili
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Engelab Ave, Tehran 14155-6451, Iran
| | - Zahra Tamiji
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Engelab Ave, Tehran 14155-6451, Iran
- Food and Drug Administration, Tehran University of Medical Sciences, Engelab Ave, Tehran 14155-6451, Iran
| | - Mehrnoosh Shirangi
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Engelab Ave, Tehran 14155-6451, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Engelab Ave, Tehran 14155-6451, Iran
| |
Collapse
|
2
|
Zohreh Mirjalili S, Chavoshi F, Amini M, ZahraTamiji, Kobarfard F, Shirangi M. Development of a high-performance liquid chromatography using rhodamine B hydrazide as the derivatization reagent for determination of β propiolactone residues in inactivated COVID-19 vaccines. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1244:124241. [PMID: 39053110 DOI: 10.1016/j.jchromb.2024.124241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/01/2024] [Accepted: 07/13/2024] [Indexed: 07/27/2024]
Abstract
β-propiolactone (BPL) is an alkylating agent used for inactivation of biological samples such as vaccines. Due to its known carcinogenic properties, complete hydrolysis of BPL is essential, and the detection of trace amounts is crucial. In this study a novel High-Performance Liquid Chromatography-Ultraviolet (HPLC-UV) method was developed. Rhodamine B hydrazide (RBH) was synthesized and utilized as a derivatizing reagent to react with BPL. The reaction was optimized in a weak acidic solution, resulting in a high yield. The separation of the RBH-derivatized BPL was achieved on a C8 column and detected by a UV detector at a wavelength of 560 nm. The method's validation demonstrated a high linearity (r2 > 0.99) over a concentration range of 0.5-50 µg/mL, with detection and quantification limits of 0.17 µg/mL and 0.5 µg/mL, respectively. The average recovery of samples was 85.20 % with a relative standard deviation (RSD) of 1.75 %. This method was successfully applied for BPL residue analysis in inactivated COVID-19 vaccines. This novel derivatization method offers a promising solution for monitoring BPL residues in the vaccine production process for quality control purposes and compliance with regulatory standards.
Collapse
Affiliation(s)
- Seyedeh Zohreh Mirjalili
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Chavoshi
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - ZahraTamiji
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Chemometrics, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Farzad Kobarfard
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrnoosh Shirangi
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; PharmaBridge, Pharmaceutical Consultancy Company, Jule Stynestraat 102 3543 DS, Utrecht, the Netherlands.
| |
Collapse
|
3
|
Schulze K, Weber U, Schuy C, Durante M, Guzmán CA. Influenza Virus Inactivated by Heavy Ion Beam Irradiation Stimulates Antigen-Specific Immune Responses. Pharmaceutics 2024; 16:465. [PMID: 38675126 PMCID: PMC11054185 DOI: 10.3390/pharmaceutics16040465] [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: 02/21/2024] [Revised: 03/22/2024] [Accepted: 03/24/2024] [Indexed: 04/28/2024] Open
Abstract
The COVID-19 pandemic has made clear the need for effective and rapid vaccine development methods. Conventional inactivated virus vaccines, together with new technologies like vector and mRNA vaccines, were the first to be rolled out. However, the traditional methods used for virus inactivation can affect surface-exposed antigen, thereby reducing vaccine efficacy. Gamma rays have been used in the past to inactivate viruses. We recently proposed that high-energy heavy ions may be more suitable as an inactivation method because they increase the damage ratio between the viral nucleic acid and surface proteins. Here, we demonstrate that irradiation of the influenza virus using heavy ion beams constitutes a suitable method to develop effective vaccines, since immunization of mice by the intranasal route with the inactivated virus resulted in the stimulation of strong antigen-specific humoral and cellular immune responses.
Collapse
Affiliation(s)
- Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Zentrum für Infektionsforschung (HZI), 38124 Braunschweig, Germany;
| | - Ulrich Weber
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany; (U.W.); (C.S.); (M.D.)
- Fachbereich Mathematik, Naturwissenschaften und Informatik, Technische Hochschule Mittelhessen, 35390 Gießen, Germany
| | - Christoph Schuy
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany; (U.W.); (C.S.); (M.D.)
| | - Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany; (U.W.); (C.S.); (M.D.)
- Institute of Condensed Matter Physics, Technische Universität Darmstadt, 64289 Darmstadt, Germany
- Department of Physics “Ettore Pancini”, University Federico II, 80138 Naples, Italy
| | - Carlos Alberto Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Zentrum für Infektionsforschung (HZI), 38124 Braunschweig, Germany;
| |
Collapse
|
4
|
He J, Kam YW. Insights from Avian Influenza: A Review of Its Multifaceted Nature and Future Pandemic Preparedness. Viruses 2024; 16:458. [PMID: 38543823 PMCID: PMC10975894 DOI: 10.3390/v16030458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 05/23/2024] Open
Abstract
Avian influenza viruses (AIVs) have posed a significant pandemic threat since their discovery. This review mainly focuses on the epidemiology, virology, pathogenesis, and treatments of avian influenza viruses. We delve into the global spread, past pandemics, clinical symptoms, severity, and immune response related to AIVs. The review also discusses various control measures, including antiviral drugs, vaccines, and potential future directions in influenza treatment and prevention. Lastly, by summarizing the insights from previous pandemic control, this review aims to direct effective strategies for managing future influenza pandemics.
Collapse
Affiliation(s)
| | - Yiu-Wing Kam
- Division of Natural and Applied Science, Duke Kunshan University, No. 8 Duke Avenue, Kunshan 215316, China;
| |
Collapse
|
5
|
Hasegawa T, Shibayama S, Osumi Y, Kato M. Optimizing virus inactivation methods for molecular detection techniques: Implications for viral protein and RNA measurements. J Virol Methods 2023; 321:114801. [PMID: 37625621 DOI: 10.1016/j.jviromet.2023.114801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023]
Abstract
To facilitate the development of effective viral detection techniques, a positive control material is required for validating their quantitative performance. Inactivated viruses serve as viable control materials, as they can be handled without the constraints of biohazard safety facilities. However, inactivation alters the structure of viral component molecules, necessitating the selection of inactivation methods that have minimal effects on the target molecules relevant to molecular detection techniques. Only a limited number of studies have investigated inactivation methods to produce viral control materials. Therefore, the aim of this study was to investigate various virus inactivation methods and evaluate their impact on molecular detection techniques, with a specific focus on viral proteins and RNA. We evaluated the effects of ultraviolet (UV) irradiation, heat, beta-propiolactone (BPL), hydrogen peroxide (H2O2), and perchloric acid (HClO4) inactivation methods to identify the most effective technique and its optimal conditions. Enzyme-linked immunosorbent assay (ELISA) and reverse transcription-digital polymerase chain reaction (RT-dPCR) were employed as model assays to assess the effects of these treatments on protein and RNA measurements. Among the evaluated methods, UV and heat treatments demonstrated minimal interference with ELISA, while heat treatment had the least impact on RT-dPCR measurements. Consequently, our findings revealed that heat inactivation holds the potential for producing inactivated viruses that can be effectively used in molecular detection techniques targeting both viral protein and RNA.
Collapse
Affiliation(s)
- Takema Hasegawa
- Bio-medical standard group, Research Institute for Material and Chemical Measurement, National Metrology Institute of Japan (NMIJ), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan.
| | - Sachie Shibayama
- Bio-medical standard group, Research Institute for Material and Chemical Measurement, National Metrology Institute of Japan (NMIJ), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Yukiko Osumi
- Bio-medical standard group, Research Institute for Material and Chemical Measurement, National Metrology Institute of Japan (NMIJ), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Megumi Kato
- Bio-medical standard group, Research Institute for Material and Chemical Measurement, National Metrology Institute of Japan (NMIJ), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| |
Collapse
|
6
|
Puente-Massaguer E, Beyer A, Loganathan M, Sapse I, Carreño JM, Bajic G, Sun W, Palese P, Krammer F. Bioprocess development for universal influenza vaccines based on inactivated split chimeric and mosaic hemagglutinin viruses. Front Bioeng Biotechnol 2023; 11:1097349. [PMID: 37342504 PMCID: PMC10277804 DOI: 10.3389/fbioe.2023.1097349] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 05/10/2023] [Indexed: 06/23/2023] Open
Abstract
Seasonal influenza viruses account for 1 billion infections worldwide every year, including 3-5 million cases of severe illness and up to 650,000 deaths. The effectiveness of current influenza virus vaccines is variable and relies on the immunodominant hemagglutinin (HA) and to a lesser extent on the neuraminidase (NA), the viral surface glycoproteins. Efficient vaccines that refocus the immune response to conserved epitopes on the HA are needed to tackle infections by influenza virus variants. Sequential vaccination with chimeric HA (cHA) and mosaic HA (mHA) constructs has proven to induce immune responses to the HA stalk domain and conserved epitopes on the HA head. In this study, we developed a bioprocess to manufacture cHA and mHA inactivated split vaccines and a method to quantify HA with a prefusion stalk based on a sandwich enzyme-linked immunosorbent assay. Virus inactivation with beta-propiolactone (βPL) and splitting with Triton X-100 yielded the highest amount of prefusion HA and enzymatically active NA. In addition, the quantity of residual Triton X-100 and ovalbumin (OVA) was reduced to very low levels in the final vaccine preparations. The bioprocess shown here provides the basis to manufacture inactivated split cHA and mHA vaccines for pre-clinical research and future clinical trials in humans, and can also be applied to produce vaccines based on other influenza viruses.
Collapse
Affiliation(s)
- Eduard Puente-Massaguer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Annika Beyer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Madhumathi Loganathan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Iden Sapse
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Goran Bajic
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Weina Sun
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Peter Palese
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
7
|
Ma Y, Steinmetz NF. Potato Virus X Inactivation and Characterization. Methods Mol Biol 2023; 2671:257-271. [PMID: 37308650 DOI: 10.1007/978-1-0716-3222-2_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The nucleoprotein components of plant viruses self-assemble into monodisperse, nanoscale structures with a high degree of symmetry and polyvalency. Of particular interest are the filamentous plant viruses which provide uniform high aspect ratio nanostructures-such structures remain challenging to obtain using purely synthetic approaches. Potato virus X (PVX) has drawn interest by the materials science community because of its filamentous structure measuring 515 × 13 nm; and both genetic engineering and chemical conjugation methods have been reported to impart new functionalities and develop PVX-based nanomaterials for applications in the health and materials sector. Toward environmentally safe materials-i.e., materials that are not infectious toward crops, such as potato, we reported methods to inactivate PVX. In this chapter, we describe the three methods to inactivate PVX and render it non-infectious toward plants, while maintaining structure and function.
Collapse
Affiliation(s)
- Yifeng Ma
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, USA
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, USA.
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
- Department of Radiology, University of California San Diego, La Jolla, CA, USA.
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, CA, USA.
- Institute for Materials Discovery and Design, University of California San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
- Center for Engineering in Cancer, Institute for Engineering in Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
8
|
Wu J, Huang W, Wang Y. Pseudotyped Viruses for the Alphavirus Chikungunya Virus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1407:299-312. [PMID: 36920704 DOI: 10.1007/978-981-99-0113-5_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Members of the genus Alphavirus are mostly mosquito-borne pathogens that cause disease in their vertebrate hosts. Chikungunya virus (CHIKV), which is one member of the genus Alphavirus [1], has been a major health problem in endemic areas since its re-emergence in 2006. CHIKV is transmitted to mammalian hosts by the Aedes mosquito, causing persistent debilitating symptoms in many cases. At present, there is no specific treatment or vaccine. Experiments involving live CHIKV need to be performed in BSL-3 facilities, which limits vaccine and drug research. The emergence of pseudotyped virus technology offered the potential for the development of a safe and effective evaluation method. In this chapter, we review the construction and application of pseudotyped CHIKVs, the findings from which have enhanced our understanding of CHIKV. This will, in turn, enable the exploration of promising therapeutic strategies in animal models, with the ultimate aim of developing effective treatments and vaccines against CHIKV and other related viruses.
Collapse
Affiliation(s)
- Jiajing Wu
- Beijing Yunling Biotechnology Co., Ltd, Beijing, China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China
| | - Youchun Wang
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China. .,Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Kunming, China.
| |
Collapse
|
9
|
Kongsomros S, Pongsakul N, Panachan J, Khowawisetsut L, Somkird J, Sangma C, Kanjanapruthipong T, Wongtrakoongate P, Chairoungdua A, Pattanapanyasat K, Newburg DS, Morrow AL, Hongeng S, Thitithanyanont A, Chutipongtanate S. Comparison of viral inactivation methods on the characteristics of extracellular vesicles from SARS-CoV-2 infected human lung epithelial cells. J Extracell Vesicles 2022; 11:e12291. [PMID: 36468940 PMCID: PMC9721205 DOI: 10.1002/jev2.12291] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 11/15/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
The interaction of SARS-CoV-2 infection with extracellular vesicles (EVs) is of particular interest at the moment. Studying SARS-CoV-2 contaminated-EV isolates in instruments located outside of the biosafety level-3 (BSL-3) environment requires knowing how viral inactivation methods affect the structure and function of extracellular vesicles (EVs). Therefore, three common viral inactivation methods, ultraviolet-C (UVC; 1350 mJ/cm2 ), β-propiolactone (BPL; 0.005%), heat (56°C, 45 min) were performed on defined EV particles and their proteins, RNAs, and function. Small EVs were isolated from the supernatant of SARS-CoV-2-infected human lung epithelial Calu-3 cells by stepwise centrifugation, ultrafiltration and qEV size-exclusion chromatography. The EV isolates contained SARS-CoV-2. UVC, BPL and heat completely abolished SARS-CoV-2 infectivity of the contaminated EVs. Particle detection by electron microscopy and nanoparticle tracking was less affected by UVC and BPL than heat treatment. Western blot analysis of EV markers was not affected by any of these three methods. UVC reduced SARS-CoV-2 spike detectability by quantitative RT-PCR and slightly altered EV-derived β-actin detection. Fibroblast migration-wound healing activity of the SARS-CoV-2 contaminated-EV isolate was only retained after UVC treatment. In conclusion, specific viral inactivation methods are compatible with specific measures in SARS-CoV-2 contaminated-EV isolates. UVC treatment seems preferable for studying functions of EVs released from SARS-CoV-2 infected cells.
Collapse
Affiliation(s)
- Supasek Kongsomros
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi HospitalMahidol UniversitySamut PrakanThailand
- Pediatric Translational Research Unit, Department of PediatricsFaculty of Medicine Ramathibodi Hospital, Mahidol UniversityBangkokThailand
- Department of Microbiology, Faculty of ScienceMahidol UniversityBangkokThailand
| | - Nutkridta Pongsakul
- Pediatric Translational Research Unit, Department of PediatricsFaculty of Medicine Ramathibodi Hospital, Mahidol UniversityBangkokThailand
| | - Jirawan Panachan
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi HospitalMahidol UniversityBangkokThailand
| | - Ladawan Khowawisetsut
- Department of Parasitology, Faculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
| | - Jinjuta Somkird
- Department of Parasitology, Faculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
| | - Chak Sangma
- Department of Chemistry, Faculty of ScienceKasetsart UniversityBangkokThailand
| | | | | | - Arthit Chairoungdua
- Department of Biochemistry, Faculty of ScienceMahidol UniversityBangkokThailand
| | - Kovit Pattanapanyasat
- Center of Excellence for Microparticle and Exosome in Diseases, Research DepartmentFaculty of Medicine Siriraj Hospital, Mahidol UniversityBangkokThailand
| | - David S. Newburg
- Division of Epidemiology, Department of Environmental and Public Health SciencesUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Ardythe L. Morrow
- Division of Epidemiology, Department of Environmental and Public Health SciencesUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Division of Infectious Diseases, Department of PediatricsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Suradej Hongeng
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi HospitalMahidol UniversityBangkokThailand
| | | | - Somchai Chutipongtanate
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi HospitalMahidol UniversitySamut PrakanThailand
- Pediatric Translational Research Unit, Department of PediatricsFaculty of Medicine Ramathibodi Hospital, Mahidol UniversityBangkokThailand
- Division of Epidemiology, Department of Environmental and Public Health SciencesUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| |
Collapse
|
10
|
Langerak T, Broekhuizen M, Unger PPA, Tan L, Koopmans M, van Gorp E, Danser AHJ, Rockx B. Transplacental Zika virus transmission in ex vivo perfused human placentas. PLoS Negl Trop Dis 2022; 16:e0010359. [PMID: 35442976 PMCID: PMC9060339 DOI: 10.1371/journal.pntd.0010359] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 05/02/2022] [Accepted: 03/25/2022] [Indexed: 11/18/2022] Open
Abstract
A Zika virus (ZIKV) infection during pregnancy can result in severe birth defects such as microcephaly. To date, it is incompletely understood how ZIKV can cross the human placenta. Furthermore, results from studies in pregnant mice and non-human primates are conflicting regarding the role of cross-reactive dengue virus (DENV) antibodies on transplacental ZIKV transmission. Elucidating how ZIKV can cross the placenta and which risk factors contribute to this is important for risk assessment and for potential intervention strategies for transplacental ZIKV transmission. In this study we use an ex vivo human placental perfusion model to study transplacental ZIKV transmission and the effect that cross-reactive DENV antibodies have on this transmission. By using this model, we demonstrate that DENV antibodies significantly increase ZIKV uptake in perfused human placentas and that this increased uptake is neonatal Fc-receptor-dependent. Furthermore, we show that cross-reactive DENV antibodies enhance ZIKV infection in term human placental explants and in primary fetal macrophages but not in primary trophoblasts. Our data supports the hypothesis that presence of cross-reactive DENV antibodies could be an important risk factor for transplacental ZIKV transmission. Furthermore, we demonstrate that the ex vivo placental perfusion model is a relevant and animal friendly model to study transplacental pathogen transmission. Zika virus is a mosquito-transmitted virus that can cause severe birth defects such as microcephaly when the infection occurs during pregnancy. Understanding how Zika virus crosses the placenta during pregnancy is important for future prevention strategies for vertical Zika virus transmission. Despite significant efforts to study this, to date it remains incompletely understood how Zika virus can cross the placenta and which risk factors contribute to this form of transmission. In this study we use an ex vivo placental perfusion model to study transplacental Zika virus transmission. The ex vivo placental perfusion model is a highly physiological and animal friendly model that mimics the in vivo conditions during pregnancy. We found that antibodies against the closely related dengue virus can significantly enhance placental uptake of Zika virus and Zika virus infection of human placental explants and fetal macrophages. These findings indicate that presence of cross-reactive dengue virus antibodies could contribute to transplacental Zika virus transmission.
Collapse
Affiliation(s)
- Thomas Langerak
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Michelle Broekhuizen
- Department of Internal Medicine, Division of Pharmacology, Erasmus Medical Center, Rotterdam, the Netherlands
- Department of Pediatrics, Division of Neonatology, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | - Lunbo Tan
- Department of Internal Medicine, Division of Pharmacology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Marion Koopmans
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Eric van Gorp
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | - A. H. Jan Danser
- Department of Internal Medicine, Division of Pharmacology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Barry Rockx
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
- * E-mail:
| |
Collapse
|
11
|
Nanometer-resolution in situ structure of the SARS-CoV-2 postfusion spike protein. Proc Natl Acad Sci U S A 2021; 118:2112703118. [PMID: 34782481 PMCID: PMC8640741 DOI: 10.1073/pnas.2112703118] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2021] [Indexed: 11/22/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a severe threat to public health and the global economy. Its spike protein is responsible for the membrane fusion and is thus a major target for vaccine and drug development. Our study presents the in situ structure of the spike protein in the postfusion state with higher resolution, giving further insights into the design of a viral entry inhibitor. Our observation of the oligomerization states of spikes on the viral membrane implies a possible mechanism of membrane fusion for viral infection. The spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mediates membrane fusion to allow entry of the viral genome into host cells. To understand its detailed entry mechanism and develop a specific entry inhibitor, in situ structural information on the SARS-CoV-2 spike protein in different states is urgent. Here, by using cryo-electron tomography, we observed both prefusion and postfusion spikes in β-propiolactone–inactivated SARS-CoV-2 virions and solved the in situ structure of the postfusion spike at nanometer resolution. Compared to previous reports, the six-helix bundle fusion core, the glycosylation sites, and the location of the transmembrane domain were clearly resolved. We observed oligomerization patterns of the spikes on the viral membrane, likely suggesting a mechanism of fusion pore formation.
Collapse
|
12
|
Volkan E. COVID-19: Structural Considerations for Virus Pathogenesis, Therapeutic Strategies and Vaccine Design in the Novel SARS-CoV-2 Variants Era. Mol Biotechnol 2021; 63:885-897. [PMID: 34145550 PMCID: PMC8213040 DOI: 10.1007/s12033-021-00353-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 06/08/2021] [Indexed: 02/08/2023]
Abstract
COVID-19 pandemic caused by SARS-CoV-2 globally impacted the humanity causing tragic outcomes; costing millions of lives, destroying economies and demolishing public health infrastructures. The emergence of vaccines using various ingenious approaches in less than a year was deemed the light at the end of the tunnel. However, recent emergence of variants of SARS-CoV-2 in several parts of the world revealed that another hurdle is ahead in the fight against COVID-19. This review will highlight how SARS-CoV-2 mutations, creating different virus variants could potentially impact virus pathogenesis as well as different therapy approaches and vaccine design.
Collapse
Affiliation(s)
- Ender Volkan
- Faculty of Pharmacy, Cyprus International University, via Mersin 10, 99258, Nicosia, Northern Cyprus, Turkey.
- Biotechnology Research Center, Cyprus International University, via Mersin 10, 99258, Nicosia, Northern Cyprus, Turkey.
| |
Collapse
|
13
|
Calvo Fernández E, Zhu LY. Racing to immunity: Journey to a COVID-19 vaccine and lessons for the future. Br J Clin Pharmacol 2021; 87:3408-3424. [PMID: 33289156 PMCID: PMC7753785 DOI: 10.1111/bcp.14686] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023] Open
Abstract
SARS-CoV-2 is the novel coronavirus behind the COVID-19 pandemic. Since its emergence, the global scientific community has mobilized to study this virus, and an overwhelming effort to identify COVID-19 treatments is currently ongoing for a variety of therapeutics and prophylactics. To better understand these efforts, we compiled a list of all COVID-19 vaccines undergoing preclinical and clinical testing using the WHO and ClinicalTrials.gov database, with details surrounding trial design and location. The most advanced vaccines are discussed in more detail, with a focus on their technology, advantages and disadvantages, as well as any available recent clinical findings. We also cover some of the primary challenges, safety concerns and public responses to COVID-19 vaccine trials, and consider what this can mean for the future. By compiling this information, we aim to facilitate a more thorough understanding of the extensive COVID-19 clinical testing vaccine landscape as it unfolds, and better highlight some of the complexities and challenges being faced by the joint effort of the scientific community in finding a prophylactic against COVID-19.
Collapse
Affiliation(s)
- Ester Calvo Fernández
- Department of Pathology and Cell BiologyColumbia University Irving Medical CenterNew YorkNYUSA
| | - Lucie Y. Zhu
- Department of Pathology and Cell BiologyColumbia University Irving Medical CenterNew YorkNYUSA
| |
Collapse
|
14
|
Gao J, Wan H, Li X, Rakic Martinez M, Klenow L, Gao Y, Ye Z, Daniels R. Balancing the influenza neuraminidase and hemagglutinin responses by exchanging the vaccine virus backbone. PLoS Pathog 2021; 17:e1009171. [PMID: 33872324 PMCID: PMC8084346 DOI: 10.1371/journal.ppat.1009171] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 04/29/2021] [Accepted: 04/05/2021] [Indexed: 01/14/2023] Open
Abstract
Virions are a common antigen source for many viral vaccines. One limitation to using virions is that the antigen abundance is determined by the content of each protein in the virus. This caveat especially applies to viral-based influenza vaccines where the low abundance of the neuraminidase (NA) surface antigen remains a bottleneck for improving the NA antibody response. Our systematic analysis using recent H1N1 vaccine antigens demonstrates that the NA to hemagglutinin (HA) ratio in virions can be improved by exchanging the viral backbone internal genes, especially the segment encoding the polymerase PB1 subunit. The purified inactivated virions with higher NA content show a more spherical morphology, a shift in the balance between the HA receptor binding and NA receptor release functions, and induce a better NA inhibitory antibody response in mice. These results indicate that influenza viruses support a range of ratios for a given NA and HA pair which can be used to produce viral-based influenza vaccines with higher NA content that can elicit more balanced neutralizing antibody responses to NA and HA. Influenza vaccines are produced on a large scale to meet the annual U.S. and global demand. To efficiently produce the required number of influenza vaccine doses, virions are commonly used as the antigen source due to their high viral protein content. A draw-back to using virions is that the final antigen composition of the vaccine is determined by the inherent properties of the vaccine virus. While this approach for influenza vaccines is beneficial for the more abundant HA antigen, it likely limits the protective response generated by the less abundant NA antigen. Our results demonstrate that the NA and HA content in vaccine viruses can be optimized by changing the internal genes of the vaccine virus, thereby preserving the surface antigens. The increase in the virion NA content that was achieved elicited higher NA antibody titres and generated more balanced neutralizing antibody responses to HA and NA. Since HA and NA neutralizing antibodies are both protective, this approach could help to improve the suboptimal efficacy of current influenza vaccines and to generate vaccines that provide broader coverage against circulating strains.
Collapse
Affiliation(s)
- Jin Gao
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Hongquan Wan
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Xing Li
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Mira Rakic Martinez
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Laura Klenow
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Yamei Gao
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Zhiping Ye
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Robert Daniels
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
- * E-mail:
| |
Collapse
|
15
|
Taddese R, Belzer C, Aalvink S, de Jonge MI, Nagtegaal ID, Dutilh BE, Boleij A. Production of inactivated gram-positive and gram-negative species with preserved cellular morphology and integrity. J Microbiol Methods 2021; 184:106208. [PMID: 33766606 DOI: 10.1016/j.mimet.2021.106208] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/26/2021] [Accepted: 03/19/2021] [Indexed: 02/07/2023]
Abstract
There are many approaches available to produce inactive bacteria by termination of growth, each with a different efficacy, impact on cell integrity, and potential for application in standardized inactivation protocols. The aim of this study was to compare these approaches and develop a standardized protocol for generation of inactivated Gram-positive and Gram-negative bacteria, yielding cells that are metabolically dead with retained cellular integrity i.e., preserving the surface and limited leakage of intracellular proteins and DNA. These inactivated bacteria are required for various applications, for instance, when investigating receptor-triggered signaling or bacterial contact-dependent analysis of cell lines requiring long incubation times. We inactivated eight different bacterial strains of different species by treatment with beta-propiolactone, ethanol, formalin, sodium hydroxide, and pasteurization. Inactivation efficacy was determined by culturing, and cell wall integrity assessed by quantifying released DNA, bacterial membrane and intracellular DNA staining, and visualization by scanning electron microscopy. Based on these results, we discuss the bacterial inactivation methods, and their advantages and disadvantages to study host-microbe interactions with inactivated bacteria.
Collapse
Affiliation(s)
- Rahwa Taddese
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University and Research, Wageningen, the Netherlands.
| | - Steven Aalvink
- Laboratory of Pediatric Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Marien I de Jonge
- Laboratory of Pediatric Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Iris D Nagtegaal
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Bas E Dutilh
- Centre for Molecular and Biomolecular Informatics, Radboud University Medical Center, Nijmegen, the Netherlands; Theoretical Biology and Bioinformatics, Utrecht University, Utrecht, the Netherlands
| | - Annemarie Boleij
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
16
|
Ke Z, Oton J, Qu K, Cortese M, Zila V, McKeane L, Nakane T, Zivanov J, Neufeldt CJ, Cerikan B, Lu JM, Peukes J, Xiong X, Kräusslich HG, Scheres SHW, Bartenschlager R, Briggs JAG. Structures and distributions of SARS-CoV-2 spike proteins on intact virions. Nature 2020; 588:498-502. [PMID: 32805734 PMCID: PMC7116492 DOI: 10.1038/s41586-020-2665-2] [Citation(s) in RCA: 806] [Impact Index Per Article: 161.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 08/10/2020] [Indexed: 02/06/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virions are surrounded by a lipid bilayer from which spike (S) protein trimers protrude1. Heavily glycosylated S trimers bind to the angiotensin-converting enzyme 2 receptor and mediate entry of virions into target cells2-6. S exhibits extensive conformational flexibility: it modulates exposure of its receptor-binding site and subsequently undergoes complete structural rearrangement to drive fusion of viral and cellular membranes2,7,8. The structures and conformations of soluble, overexpressed, purified S proteins have been studied in detail using cryo-electron microscopy2,7,9-12, but the structure and distribution of S on the virion surface remain unknown. Here we applied cryo-electron microscopy and tomography to image intact SARS-CoV-2 virions and determine the high-resolution structure, conformational flexibility and distribution of S trimers in situ on the virion surface. These results reveal the conformations of S on the virion, and provide a basis from which to understand interactions between S and neutralizing antibodies during infection or vaccination.
Collapse
Affiliation(s)
- Zunlong Ke
- Structural Studies Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Joaquin Oton
- Structural Studies Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Kun Qu
- Structural Studies Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Mirko Cortese
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, Heidelberg, Germany
| | - Vojtech Zila
- Department of Infectious Diseases, Virology, Heidelberg University, Heidelberg, Germany
| | - Lesley McKeane
- Visual Aids Department, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Takanori Nakane
- Structural Studies Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Jasenko Zivanov
- Structural Studies Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Christopher J Neufeldt
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, Heidelberg, Germany
| | - Berati Cerikan
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, Heidelberg, Germany
| | - John M Lu
- Structural Studies Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Julia Peukes
- Structural Studies Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Xiaoli Xiong
- Structural Studies Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Hans-Georg Kräusslich
- Department of Infectious Diseases, Virology, Heidelberg University, Heidelberg, Germany
- German Center for Infection Research, Heidelberg Partner Site, Heidelberg, Germany
| | - Sjors H W Scheres
- Structural Studies Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, Heidelberg, Germany
- German Center for Infection Research, Heidelberg Partner Site, Heidelberg, Germany
- Division of Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - John A G Briggs
- Structural Studies Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK.
| |
Collapse
|
17
|
Rezinciuc S, Bezavada L, Bahadoran A, Duan S, Wang R, Lopez-Ferrer D, Finkelstein D, McGargill MA, Green DR, Pasa-Tolic L, Smallwood HS. Dynamic metabolic reprogramming in dendritic cells: An early response to influenza infection that is essential for effector function. PLoS Pathog 2020; 16:e1008957. [PMID: 33104753 PMCID: PMC7707590 DOI: 10.1371/journal.ppat.1008957] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/01/2020] [Accepted: 09/03/2020] [Indexed: 01/19/2023] Open
Abstract
Infection with the influenza virus triggers an innate immune response that initiates the adaptive response to halt viral replication and spread. However, the metabolic response fueling the molecular mechanisms underlying changes in innate immune cell homeostasis remain undefined. Although influenza increases parasitized cell metabolism, it does not productively replicate in dendritic cells. To dissect these mechanisms, we compared the metabolism of dendritic cells to that of those infected with active and inactive influenza A virus and those treated with toll-like receptor agonists. Using quantitative mass spectrometry, pulse chase substrate utilization assays and metabolic flux measurements, we found global metabolic changes in dendritic cells 17 hours post infection, including significant changes in carbon commitment via glycolysis and glutaminolysis, as well as mitochondrial respiration. Influenza infection of dendritic cells led to a metabolic phenotype distinct from that induced by TLR agonists, with significant resilience in terms of metabolic plasticity. We identified c-Myc as one transcription factor modulating this response. Restriction of c-Myc activity or mitochondrial substrates significantly changed the immune functions of dendritic cells, such as reducing motility and T cell activation. Transcriptome analysis of inflammatory dendritic cells isolated following influenza infection showed similar metabolic reprogramming occurs in vivo. Thus, early in the infection process, dendritic cells respond with global metabolic restructuring, that is present in inflammatory lung dendritic cells after infection, and this is important for effector function. These findings suggest metabolic switching in dendritic cells plays a vital role in initiating the immune response to influenza infection. Dendritic cells are critical in mounting an effective immune response to influenza infection by initiating the immune response to influenza and activating the adaptive response to mediate viral clearance and manifest immune memory for protection against subsequent infections. We found dendritic cells undergo a profound metabolic shift after infection. They alter the concentration and location of hundreds of proteins, including c-Myc, facilitating a shift to a highly glycolytic phenotype that is also flexible in terms of fueling respiration. Nonetheless, we found limiting access to specific metabolic pathways or substrates diminished key immune functions. We previously described an immediate, fixed hypermetabolic state in infected respiratory epithelial cells. Here we present data indicating the metabolic response of dendritic cells is increased yet flexible, distinct from what we previously showed for epithelial cells. Additionally, we demonstrate dendritic cells tailor their metabolic response to the pathogen or TLR stimulus. This metabolic reprogramming occurs rapidly in vitro and is sustained in inflammatory dendritic cells in vivo for at least 9 days following influenza infection. These studies introduce the possibility of modulating the immune response to viral infection using customized metabolic therapy to enhance or diminish the function of specific cells.
Collapse
Affiliation(s)
- Svetlana Rezinciuc
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Lavanya Bezavada
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Azadeh Bahadoran
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Susu Duan
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Ruoning Wang
- Center for Childhood Cancer and Blood Disease, The Research Institute at Nationwide Children's Hospital, The Ohio State University School of Medicine, Columbus, Ohio, United States of America
| | - Daniel Lopez-Ferrer
- Chromatography and Mass Spectrometry Division, Thermo Fisher Scientific, CA, United States of America
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Maureen A. McGargill
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Douglas R. Green
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Ljiljana Pasa-Tolic
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington, United States of America
| | - Heather S. Smallwood
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
18
|
Zhao F, Liu L, Xu M, Shu X, Zheng L, Wei Z. Assessments of different inactivating reagents in formulating transmissible gastroenteritis virus vaccine. Virol J 2020; 17:163. [PMID: 33097081 PMCID: PMC7582447 DOI: 10.1186/s12985-020-01433-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/12/2020] [Indexed: 11/21/2022] Open
Abstract
Background Transmissible gastroenteritis virus (TGEV) causes enteric infection in piglets, characterized by vomiting, severe diarrhea and dehydration, and the mortality in suckling piglets is often high up to 100%. Vaccination is an effective measure to control the disease caused by TGEV. Methods In this study, cell-cultured TGEV HN-2012 strain was inactivated by formaldehyde (FA), β-propiolactone (BPL) or binaryethylenimine (BEI), respectively. Then the inactivated TGEV vaccine was prepared with freund's adjuvant, and the immunization effects were evaluated in mice. The TGEV-specific IgG level was detected by ELISA. The positive rates of CD4+, CD8+, CD4+IFN-γ+, CD4+IL-4+ T lymphocytes were detected by flow cytometry assay. Lymphocyte proliferation assay and gross pathology and histopathology examination were also performed to assess the three different inactivating reagents in formulating TGEV vaccine. Results The results showed that the TGEV-specific IgG level in FA group (n = 17) was earlier and stronger, while the BEI group produced much longer-term IgG level. The lymphocyte proliferation test demonstrated that the BEI group had a stronger ability to induce spleen lymphocyte proliferation. The positive rates of CD4+ and CD8+ T lymphocyte subsets of peripheral blood lymphocyte in BEI group was higher than that in FA group and BPL groups by flow cytometry assay. The positive rate of CD4+IFN-γ+ T lymphocyte subset was the highest in the BPL group, and the positive rate of CD4+IL-4+ T lymphocyte subset was the highest in the FA group. There were no obvious pathological changes in the vaccinated mice and the control group after the macroscopic and histopathological examination. Conclusions These results indicated that all the three experimental groups could induce cellular and humoral immunity, and the FA group had the best humoral immunity effect, while the BEI group showed its excellent cellular immunity effect.
Collapse
Affiliation(s)
- Fujie Zhao
- The College of Veterinary Medicine, Henan Agricultural University, Nongye Road 63#, Zhengzhou, 450002, Henan Province, People's Republic of China
| | - Lintao Liu
- The College of Veterinary Medicine, Henan Agricultural University, Nongye Road 63#, Zhengzhou, 450002, Henan Province, People's Republic of China
| | - Menglong Xu
- The College of Veterinary Medicine, Henan Agricultural University, Nongye Road 63#, Zhengzhou, 450002, Henan Province, People's Republic of China
| | - Xiangli Shu
- The College of Veterinary Medicine, Henan Agricultural University, Nongye Road 63#, Zhengzhou, 450002, Henan Province, People's Republic of China
| | - Lanlan Zheng
- The College of Veterinary Medicine, Henan Agricultural University, Nongye Road 63#, Zhengzhou, 450002, Henan Province, People's Republic of China.
| | - Zhanyong Wei
- The College of Veterinary Medicine, Henan Agricultural University, Nongye Road 63#, Zhengzhou, 450002, Henan Province, People's Republic of China. .,Key Laboratory for Animal-Derived Food Safety of Henan Province, Zhengzhou, 450002, Henan, China.
| |
Collapse
|
19
|
Sitaras I, Spackman E, de Jong MCM, Parris DJ. Selection and antigenic characterization of immune-escape mutants of H7N2 low pathogenic avian influenza virus using homologous polyclonal sera. Virus Res 2020; 290:198188. [PMID: 33045306 DOI: 10.1016/j.virusres.2020.198188] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 09/18/2020] [Accepted: 10/05/2020] [Indexed: 01/24/2023]
Abstract
Understanding the dynamics of the selection of influenza A immune escape variants by serum antibody is critical for designing effective vaccination programs for animals, especially poultry where large populations have a short generation time and may be vaccinated with high frequency. In this report, immune-escape mutants of A/turkey/New York/4450/1994 H7N2 low pathogenic avian influenza virus, were selected by serially passaging the virus in the presence of continuously increasing concentrations of homologous chicken polyclonal sera. Amino acid mutations were identified by sequencing the parental hemagglutinin (HA) gene and every 10 passages by both Sanger and deep sequencing, and the antigenic distance of the mutants to the parent strain was determined. Progressively, a total of five amino acid mutations were observed over the course of 30 passages. Based on their absence from the parental virus with deep sequencing, the mutations appear to have developed de novo. The antigenic distance between the selected mutants and the parent strain increased as the number of amino acid mutations accumulated and the concentration of antibodies had to be periodically increased to maintain the same reduction in virus titer during selection. This selection system demonstrates how H7 avian influenza viruses behave under selection with homologous sera, and provides a glimpse of their evolutionary dynamics, which can be applied to developing vaccination programs that maximize the effectiveness of a vaccine over time.
Collapse
Affiliation(s)
- Ioannis Sitaras
- Exotic and Emerging Avian Viral Diseases Unit, Southeast Poultry Research Laboratory, United States National Poultry Research Center, United States Department of Agriculture, Agricultural Research Service, 934 College Station Road, Athens, GA, 30605, USA
| | - Erica Spackman
- Exotic and Emerging Avian Viral Diseases Unit, Southeast Poultry Research Laboratory, United States National Poultry Research Center, United States Department of Agriculture, Agricultural Research Service, 934 College Station Road, Athens, GA, 30605, USA.
| | - Mart C M de Jong
- Quantitative Veterinary Epidemiology, Wageningen University and Research, Droevendaalsesteeg 1, 6708PB, Wageningen, The Netherlands
| | - D Joshua Parris
- Exotic and Emerging Avian Viral Diseases Unit, Southeast Poultry Research Laboratory, United States National Poultry Research Center, United States Department of Agriculture, Agricultural Research Service, 934 College Station Road, Athens, GA, 30605, USA
| |
Collapse
|
20
|
Abstract
![]()
The
discovery that β-propiolactone (BPL), once a commercially
important chemical, causes various tumors in experimental animals
has led to a significant decrease in its use. However, owing to its
efficacy this possible human carcinogen remains to be utilized in
vaccines for inactivation of viruses. The focus of the current study
was to uncover the mechanisms of β-propiolactone reactions with
both nucleobases and glutathione (GSH) through computer simulations
based on quantum chemical methods. Our results, in accordance with
in vitro studies, show that among all nucleobases guanine most readily
forms adducts with BPL through SN2 reaction mechanism.
Acquired activation energies with incorporated solvent effects reveal
that alkylation represents an energetically more favorable reaction
than acylation for all nucleobases. Comparison of activation free
energies of glutathione and guanine reactions with BPL suggest that
glutathione may represent an efficient natural scavenger of BPL. Therefore,
glutathione present in the organism may provide protection to the
DNA and thus prevent BPL’s genotoxicity, mutagenicity, and
possibly even carcinogenicity.
Collapse
Affiliation(s)
- Eva Španinger
- Laboratory of Physical Chemistry and Chemical Thermodynamics, Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova ulica 17, SI-2000 Maribor, Slovenia
| | - Urban Bren
- Laboratory of Physical Chemistry and Chemical Thermodynamics, Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova ulica 17, SI-2000 Maribor, Slovenia
| |
Collapse
|
21
|
Furlan V, Bren U. Protective Effects of [6]-Gingerol Against Chemical Carcinogens: Mechanistic Insights. Int J Mol Sci 2020; 21:E695. [PMID: 31973096 PMCID: PMC7037038 DOI: 10.3390/ijms21030695] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/18/2020] [Accepted: 01/20/2020] [Indexed: 12/28/2022] Open
Abstract
[6]-Gingerol from ginger has received considerable attention as a potential cancer therapeutic agent because of its chemopreventive and chemotherapeutic effects, as well as its safety. In the current study, we examined [6]-gingerol as a natural scavenger of nine ultimate chemical carcinogens to which we are frequently exposed: glycidamide, styrene oxide, aflatoxin B1 exo-8,9-epoxide, β-propiolactone, ethylene oxide, propylene oxide, 2-cyanoethylene oxide, chloroethylene oxide, and vinyl carbamate epoxide. To evaluate [6]-gingerol efficacy, we expanded our research with the examination of glutathione-the strongest natural scavenger in human cells. The corresponding activation free energies were calculated using Hartree-Fock method with three flexible basis sets and two implicit solvation models. According to our results, [6]-gingerol proves to be an extremely effective scavenger of chemical carcinogens of the epoxy type. On the other hand, with the exception of aflatoxin B1 exo-8,9-epoxide, glutathione represents a relatively poor scavenger, whose efficacy could be augmented by [6]-gingerol. Moreover, our quantum mechanical study of the alkylation reactions of chemical carcinogens with [6]-gingerol and glutathione provide valuable insights in the reaction mechanisms and the geometries of the corresponding transition states. Therefore, we strongly believe that our research forms a solid basis for further computational, experimental and clinical studies of anticarcinogenic properties of [6]-gingerol as well as for the development of novel chemoprophylactic dietary supplements. Finally, the obtained results also point to the applicability of quantum chemical methods to studies of alkylation reactions related to chemical carcinogenesis.
Collapse
Affiliation(s)
- Veronika Furlan
- Faculty of Chemistry and Chemical Technology, University of Maribor, Smetanova 17, SI-2000 Maribor, Slovenia;
| | - Urban Bren
- Faculty of Chemistry and Chemical Technology, University of Maribor, Smetanova 17, SI-2000 Maribor, Slovenia;
- National Institute of Chemistry, Hajdrihova 19, SI-1001 Ljubljana, Slovenia
| |
Collapse
|
22
|
Wiehe A, O'Brien JM, Senge MO. Trends and targets in antiviral phototherapy. Photochem Photobiol Sci 2019; 18:2565-2612. [PMID: 31397467 DOI: 10.1039/c9pp00211a] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Photodynamic therapy (PDT) is a well-established treatment option in the treatment of certain cancerous and pre-cancerous lesions. Though best-known for its application in tumor therapy, historically the photodynamic effect was first demonstrated against bacteria at the beginning of the 20th century. Today, in light of spreading antibiotic resistance and the rise of new infections, this photodynamic inactivation (PDI) of microbes, such as bacteria, fungi, and viruses, is gaining considerable attention. This review focuses on the PDI of viruses as an alternative treatment in antiviral therapy, but also as a means of viral decontamination, covering mainly the literature of the last decade. The PDI of viruses shares the general action mechanism of photodynamic applications: the irradiation of a dye with light and the subsequent generation of reactive oxygen species (ROS) which are the effective phototoxic agents damaging virus targets by reacting with viral nucleic acids, lipids and proteins. Interestingly, a light-independent antiviral activity has also been found for some of these dyes. This review covers the compound classes employed in the PDI of viruses and their various areas of use. In the medical area, currently two fields stand out in which the PDI of viruses has found broader application: the purification of blood products and the treatment of human papilloma virus manifestations. However, the PDI of viruses has also found interest in such diverse areas as water and surface decontamination, and biosafety.
Collapse
Affiliation(s)
- Arno Wiehe
- biolitec research GmbH, Otto-Schott-Str. 15, 07745 Jena, Germany. and Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Jessica M O'Brien
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James's Hospital, Dublin 8, Ireland.
| | - Mathias O Senge
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James's Hospital, Dublin 8, Ireland.
| |
Collapse
|
23
|
Sabbaghi A, Miri SM, Keshavarz M, Zargar M, Ghaemi A. Inactivation methods for whole influenza vaccine production. Rev Med Virol 2019; 29:e2074. [PMID: 31334909 DOI: 10.1002/rmv.2074] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/16/2019] [Accepted: 06/19/2019] [Indexed: 12/16/2022]
Abstract
Despite tremendous efforts toward vaccination, influenza remains an ongoing global threat. The induction of strain-specific neutralizing antibody responses is a common phenomenon during vaccination with the current inactivated influenza vaccines, so the protective effect of these vaccines is mostly strain-specific. There is an essential need for the development of next-generation vaccines, with a broad range of immunogenicity against antigenically drifted or shifted influenza viruses. Here, we evaluate the potential of whole inactivated vaccines, based on chemical and physical methods, as well as new approaches to generate cross-protective immune responses. We also consider the mechanisms by which some of these vaccines may induce CD8+ T-cells cross-reactivity with different strains of influenza. In this review, we have focused on conventional and novel methods for production of whole inactivated influenza vaccine. As well as chemical modification, using formaldehyde or β-propiolactone and physical manipulation by ultraviolet radiation or gamma-irradiation, novel approaches, including visible ultrashort pulsed laser, and low-energy electron irradiation are discussed. These two latter methods are considered to be attractive approaches to design more sophisticated vaccines, due to their ability to maintain most of the viral antigenic properties during inactivation and potential to produce cross-protective immunity. However, further studies are needed to validate them before they can replace traditional methods for vaccine manufacturing.
Collapse
Affiliation(s)
- Ailar Sabbaghi
- Department of Microbiology, Qom Branch, Islamic Azad University, Qom, Iran.,Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran
| | | | - Mohsen Keshavarz
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohsen Zargar
- Department of Microbiology, Qom Branch, Islamic Azad University, Qom, Iran
| | - Amir Ghaemi
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
24
|
Potency test to discriminate between differentially over-inactivated rabies vaccines: Agreement between the NIH assay and a G-protein based ELISA. Biologicals 2019; 60:49-54. [DOI: 10.1016/j.biologicals.2019.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 05/13/2019] [Accepted: 05/14/2019] [Indexed: 11/18/2022] Open
|
25
|
Adi AAAM, Astawa INM, Putra IGAA. The efficacy of binary ethylenimine-inactivated vaccines of Gianyar-1/AK/2014 virulent strain in protecting chickens against Tabanan-1/ARP/2017 virulent Newcastle disease virus isolates. Vet World 2019; 12:758-764. [PMID: 31439989 PMCID: PMC6661483 DOI: 10.14202/vetworld.2019.758-764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 04/11/2019] [Indexed: 11/16/2022] Open
Abstract
AIM This study aimed to prepare binary ethylenimine (BEI)-inactivated virulent Newcastle disease virus (NDV) vaccine and to examine their ability to induce a protective antibody response in commercial chickens. MATERIALS AND METHODS A virulent NDV field isolate Gianyar-1/AK/2014 was propagated in chicken-embryonated eggs and was then inactivated with BEI at a concentration of 4 mM. Three groups of chickens with low-level (2 log2 hemagglutination inhibition [HI] units) maternally derived antibodies against NDV were then immunized with the BEI-inactivated vaccine. A commercial live vaccine (LaSota strain) was used as positive control, and phosphate-buffered saline (PBS) was used as negative control. A challenge experiment with a virulent NDV of Tabanan-1/ARP/2017 was performed at 3 weeks post-vaccination. RESULTS At 2 weeks post-immunization, the mean titers of antibodies against NDV in serum samples of chickens immunized with 0.2 mL of BEI-inactivated NDV (Group I), with live commercial NDV vaccine (Group II) and with PBS (Group III) were 3±0.94 log2 HI units, 4.9±0.99 log2 HI unit, and 0.0±0.0 HI units, respectively. At week 3 post-immunization, the mean titers of the antibodies for the three groups were 5±1.09 log2 HI units, 6.9±0.32 log2 HI units, and 0.00 HI units, respectively. The antibody titer induced by inactivated NDV Gianyar-1/AK/2014 isolates examined at 2 and 3 weeks post-vaccination was still at a significantly (p<0.01) lower level as compared to those induced by commercial life vaccine. However, the challenge test with virulent NDV of Tabanan 1/ARP/2017 isolates showed that all immunized chickens (Group I and II) survived without exhibiting any clinical sign post-challenge with the protection rates of 100%, whereas all chickens injected with PBS (Group III) died with clinical signs of ND. CONCLUSION This finding shows that the BEI-inactivated vaccines prepared using virulent NDV of Gianyar-1/AK/2014 strain was able to induce protective antibody response in chickens but still at a lower level than those induce by commercial live NDV vaccine.
Collapse
Affiliation(s)
- Anak Agung Ayu Mirah Adi
- Laboratory of Veterinary Pathology, Faculty of Veterinary Medicine, Udayana University, Kampus Sudirman, Jalan PB Sudirman, Denpasar, Bali, Indonesia
| | - I Nyoman Mantik Astawa
- Laboratory of Veterinary Virology, Faculty of Veterinary Medicine Udayana University, Kampus Sudirman, Jalan PB Sudirman, Denpasar, Bali, Indonesia
| | - I Gusti Agung Arta Putra
- Laboratory of Animal Anatomy and Physiology, Faculty of Animal Husbandry, Udayana University, Kampus Bukit, Jimbaran, Badung, Bali, Indonesia
| |
Collapse
|
26
|
Keshwara R, Shiels T, Postnikova E, Kurup D, Wirblich C, Johnson RF, Schnell MJ. Rabies-based vaccine induces potent immune responses against Nipah virus. NPJ Vaccines 2019; 4:15. [PMID: 31016033 PMCID: PMC6465360 DOI: 10.1038/s41541-019-0109-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 03/04/2019] [Indexed: 12/25/2022] Open
Abstract
Nipah Virus (NiV) is a re-emerging zoonotic pathogen in the genus Henipavirus of the Paramyxoviridae family of viruses. NiV is endemic to Bangladesh and Malaysia and is highly fatal to both livestock and humans (human case fatality rate = 74.5%). Currently, there is no approved vaccine against NiV on the market. The goal of this study was to use a recombinant RABV vector expressing NiV glycoprotein (NiV G) to develop a bivalent candidate vaccine against NiV disease and rabies virus (RABV) disease, which is also a significant health burden in the regions where NiV is endemic. The rabies vector is a well-established vaccine strain that lacks neurovirulence and can stably expresses foreign antigens that are immunogenic in various animal models. Mice inoculated intranasally with the live recombinant RABV/NiV vaccine (NIPARAB) showed no signs of disease. To test the immunogenicity of the vaccine candidate, groups of C57BL/6 mice were immunized intramuscularly with a single dose of live vaccine particles or two doses of chemically inactivated viral particles. Both vaccination groups showed NiV G-specific seroconversion, and the inactivated (INAC) vaccine group yielded higher titers of NiV G-specific antibodies. Furthermore, cross-reactivity of NiV G-specific immune sera against Hendra virus (HeV), was confirmed by immunofluorescence (IF) and indirect ELISA against soluble recombinant HeV glycoprotein (HeV G). Both live and killed vaccines induced neutralizing antibodies. These results indicate that NIPARAB may be used as a killed virus vaccine to protect humans against NiV and RABV, and possibly as a preventative measure against HeV as well.
Collapse
Affiliation(s)
- Rohan Keshwara
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Thomas Shiels
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Elena Postnikova
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, MD 21702 USA
| | - Drishya Kurup
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Christoph Wirblich
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Reed F. Johnson
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Matthias J. Schnell
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107 USA
- Jefferson Vaccine Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107 USA
| |
Collapse
|
27
|
Herrera-Rodriguez J, Signorazzi A, Holtrop M, de Vries-Idema J, Huckriede A. Inactivated or damaged? Comparing the effect of inactivation methods on influenza virions to optimize vaccine production. Vaccine 2019; 37:1630-1637. [PMID: 30765167 PMCID: PMC7115651 DOI: 10.1016/j.vaccine.2019.01.086] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 01/22/2019] [Accepted: 01/28/2019] [Indexed: 11/27/2022]
Abstract
β-propiolactone (BPL) and formaldehyde (FA) were used to inactivate several influenza virus strains. BPL abolished the infectivity, FA was unable to completely inactivate the virus. All methods damaged the binding and fusion capacity; BPL caused greater loss than FA. FA treatments caused the highest reduction in TLR-7 stimulation. All the observed effects were strain-dependent.
The vast majority of commercially available inactivated influenza vaccines are produced from egg-grown or cell-grown live influenza virus. The first step in the production process is virus inactivation with β-propiolactone (BPL) or formaldehyde (FA). Recommendations for production of inactivated vaccines merely define the maximal concentration for both reagents, leaving the optimization of the process to the manufacturers. We assessed the effect of inactivation with BPL and FA on 5 different influenza virus strains. The properties of the viral formulation, such as successful inactivation, preservation of hemagglutinin (HA) binding ability, fusion capacity and the potential to stimulate a Toll-like receptor 7 (TLR7) reporter cell line were then assessed and compared to the properties of the untreated virus. Inactivation with BPL resulted in undetectable infectivity levels, while FA-treated virus retained very low infectious titers. Hemagglutination and fusion ability were highly affected by those treatments that conferred higher inactivation, with BPL-treated virus binding and fusing at a lower degree compared to FA-inactivated samples. On the other hand, BPL-inactivated virus induced higher levels of activation of TLR7 than FA-inactivated virus. The alterations caused by BPL or FA treatments were virus strain dependent. This data shows that the inactivation procedures should be tailored on the virus strain, and that many other elements beside the concentration of the inactivating agent, such as incubation time and temperature, buffer and virus concentration, have to be defined to achieve a functional product.
Collapse
Affiliation(s)
- José Herrera-Rodriguez
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Aurora Signorazzi
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Marijke Holtrop
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jacqueline de Vries-Idema
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Anke Huckriede
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
28
|
Theillet G, Grard G, Galla M, Maisse C, Enguehard M, Cresson M, Dalbon P, Leparc-Goffart IL, Bedin F. Detection of chikungunya virus-specific IgM on laser-cut paper-based device using pseudo-particles as capture antigen. J Med Virol 2019; 91:899-910. [PMID: 30734316 DOI: 10.1002/jmv.25420] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/21/2019] [Accepted: 01/23/2019] [Indexed: 01/11/2023]
Abstract
The incidence of arbovirus infections has increased dramatically in recent decades, affecting hundreds of millions of people each year. The Togaviridae family includes the chikungunya virus (CHIKV), which is typically transmitted by Aedes mosquitoes and causes a wide range of symptoms from flu-like fever to severe arthralgia. Although conventional diagnostic tests can provide early diagnosis of CHIKV infections, access to these tests is often limited in developing countries. Consequently, there is an urgent need to develop efficient, affordable, simple, rapid, and robust diagnostic tools that can be used in point-of-care settings. Early diagnosis is crucial to improve patient management and to reduce the risk of complications. A glass-fiber laser-cut microfluidic device (paper-based analytical device [PAD]) was designed and evaluated in a proof of principle context, for the analysis of 30 µL of patient serum. Biological raw materials used for the functionalization of the PAD were first screened by MAC-ELISA (IgM capture enzyme-linked immunosorbent assay) for CHIKV Immunoglobulin M (IgM) capture and then evaluated on the PAD using various human samples. Compared with viral lysate traditionally used for chikungunya (CHIK) serology, CHIKV pseudo-particles (PPs) have proven to be powerful antigens for specific IgM capture. The PAD was able to detect CHIKV IgM in human sera in less than 10 minutes. Results obtained in patient sera showed a sensitivity of 70.6% and a specificity of around 98%. The PAD showed few cross-reactions with other tropical viral diseases. The PAD could help health workers in the early diagnosis of tropical diseases such as CHIK, which require specific management protocols in at-risk populations.
Collapse
Affiliation(s)
- Gerald Theillet
- bioMérieux, Innovations New Immuno-Concepts department, Chemin de l'Orme, Marcy-l'Etoile, France.,Unité des Virus Emergents (UVE: Aix-Marseille Univ, IRD 190, Inserm 1207, IHU Méditerranée Infection), Marseille, France
| | - Gilda Grard
- Unité des Virus Emergents (UVE: Aix-Marseille Univ, IRD 190, Inserm 1207, IHU Méditerranée Infection), Marseille, France.,IRBA, Unité de virologie, CNR des Arbovirus, HIA Laveran, Marseille, France
| | - Mathilde Galla
- Unité des Virus Emergents (UVE: Aix-Marseille Univ, IRD 190, Inserm 1207, IHU Méditerranée Infection), Marseille, France.,IRBA, Unité de virologie, CNR des Arbovirus, HIA Laveran, Marseille, France
| | - Carine Maisse
- Infections Virales et Pathologie Comparée, UMR754, INRA, Univ Claude Bernard Lyon1, Lyon, France
| | - Margot Enguehard
- Ecologie Microbienne CNRS UMR 5557, INRA UMR1418, Villeurbanne, France.,CAS Key Laboratory of Molecular Virology and Immunology, Unit of Interspecies transmission of arboviruses and antivirals, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Marie Cresson
- CAS Key Laboratory of Molecular Virology and Immunology, Unit of Interspecies transmission of arboviruses and antivirals, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,IVPC UMR754, INRA, Univ Lyon, Université Claude Bernard Lyon 1, EPHE, PSL Research University, Lyon, France
| | - Pascal Dalbon
- bioMérieux, Innovations New Immuno-Concepts department, Chemin de l'Orme, Marcy-l'Etoile, France
| | - Isabelle Leparc Leparc-Goffart
- Unité des Virus Emergents (UVE: Aix-Marseille Univ, IRD 190, Inserm 1207, IHU Méditerranée Infection), Marseille, France.,IRBA, Unité de virologie, CNR des Arbovirus, HIA Laveran, Marseille, France
| | - Frederic Bedin
- bioMérieux, Innovations New Immuno-Concepts department, Chemin de l'Orme, Marcy-l'Etoile, France
| |
Collapse
|
29
|
Characterisation of the antigenic epitopes in the subunit 2 haemagglutinin of avian influenza virus H5N1. Arch Virol 2018; 163:2199-2212. [DOI: 10.1007/s00705-018-3896-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/19/2018] [Indexed: 01/21/2023]
|
30
|
Sibanda T, Selvarajan R, Tekere M, Nyoni H, Meddows-Taylor S. Potential biotechnological capabilities of cultivable mycobiota from carwash effluents. Microbiologyopen 2017; 6. [PMID: 28714266 PMCID: PMC5635173 DOI: 10.1002/mbo3.498] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/12/2017] [Accepted: 04/25/2017] [Indexed: 12/22/2022] Open
Abstract
Urban life has created man‐made extreme environments like carwashes. These environments have, however, not been sufficiently explored for mycobiota that can be sources of biotechnologically useful products, as has been the case with natural extreme environments. Using a combination of culture and molecular techniques, fungi from carwash effluents was characterized for production of lipase and cellulase enzymes, nonpolar and polar biotechnologically relevant secondary metabolites and hydrocarbon utilization. The isolated fungal strains belonged to the genera Alternaria, Cladosporium, Penicillium, Peyronellaea, Rhizopus, Spegazzinia, Trichoderma, Ulocladium and Yarrowia. Sixty‐six percent (66%) of the fungal isolates were found to be able to metabolize naphthalene and benzanthracene, showing potential for application in bioremediation of hydrocarbon polluted sites. Lipase production by the isolates Penicillium sp. BPS3 (2.61 U/ml), Trichoderma sp. BPS9 (2.01 U/ml), Rhizopus sp. CAL1 (2.05 U/ml), Penicillium sp. PCW1 (2.99 U/ml) and Penicillium sp. SAS1 (2.16 U/ml) compared well with previously recorded lipase production levels by other fungi. The highest producers of cellulase were Penicillium sp. SAS1 (12.10 U/ml), Peyronella sp. CAW5 (4.49 U/ml) and Cladosporium sp. SAS3 (4.07 U/ml), although these activities were lower than previously reported levels. GC‐MS analysis of the fungal secondary metabolites resulted in identification of 572 compounds, including azulene, methanamine, N‐pentylidene, metoclopramide, and mepivacaine while compounds determined by UHPLC‐MS included 10‐undecen‐1‐ol, piquerol A, 10‐undecyn‐1‐ol, cyclo(leucylprolyl) and rac‐etomidate. These compounds were previously determined to have various activities including anticancer, antibacterial, antifungal, antihypertensive, antidiabetic and anti‐inflammatory properties. The study demonstrated that fungi from carwash effluents are natural sources of some biotechnologically important products.
Collapse
Affiliation(s)
- Timothy Sibanda
- Department of Environmental Sciences, College of Agriculture and Environmental Sciences, Florida, South Africa
| | - Ramganesh Selvarajan
- Department of Environmental Sciences, College of Agriculture and Environmental Sciences, Florida, South Africa
| | - Memory Tekere
- Department of Environmental Sciences, College of Agriculture and Environmental Sciences, Florida, South Africa
| | - Hlengilizwe Nyoni
- Department of Nanotechnology and Water Sustainability, College of Science, Engineering and Technology, Florida, South Africa
| | | |
Collapse
|
31
|
Lone NA, Spackman E, Kapczynski D. Immunologic evaluation of 10 different adjuvants for use in vaccines for chickens against highly pathogenic avian influenza virus. Vaccine 2017; 35:3401-3408. [DOI: 10.1016/j.vaccine.2017.05.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/17/2017] [Accepted: 05/03/2017] [Indexed: 12/16/2022]
|
32
|
Beta-Propiolactone Inactivation of Coxsackievirus A16 Induces Structural Alteration and Surface Modification of Viral Capsids. J Virol 2017; 91:JVI.00038-17. [PMID: 28148783 DOI: 10.1128/jvi.00038-17] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 01/12/2017] [Indexed: 11/20/2022] Open
Abstract
Beta-propiolactone (BPL) is an inactivating agent that is widely used in the vaccine industry. However, its effects on vaccine protein antigens and its mechanisms of action remain poorly understood. Here we present cryo-electron microscopy (cryo-EM) structures of BPL-treated coxsackievirus A16 (CVA16) mature virions and procapsids at resolutions of 3.9 Å and 6.5 Å, respectively. Notably, both particles were found to adopt an expanded conformation resembling the 135S-like uncoating intermediate, with characteristic features including an opened 2-fold channel, the externalization of the N terminus of VP1 capsid protein, and the absence of pocket factor. However, major neutralizing epitopes are very well preserved on these particles. Further biochemical analyses revealed that BPL treatment impairs the abilities of CVA16 particles to bind to the attachment receptor heparan sulfate and to a conformation-dependent monoclonal antibody in a BPL dose-dependent manner, indicating that BPL is able to modify surface-exposed amino acid residues. Taken together, our results demonstrate that BPL treatment may induce alteration of the overall structure and surface properties of a nonenveloped viral capsid, thus revealing a novel mode of action of BPL.IMPORTANCE Beta-propiolactone (BPL) is commonly used as an inactivating reagent to produce viral vaccines. It is recognized that BPL inactivates viral infectivity through modification of viral nucleic acids. However, its effect on viral proteins remains largely unknown. Here, we present high-resolution cryo-EM structures of BPL-treated coxsackievirus A16 (CVA16) mature virions and procapsids, which reveals an expanded overall conformation and characteristic features that are typical for the 135S-like uncoating intermediate. We further show that the BPL concentration affects the binding of inactivated CVA16 particles to their receptor/antibody. Thus, BPL treatment can alter the overall structure and surface properties of viral capsids, which may lead to antigenic and immunogenic variations. Our findings provide important information for future development of BPL-inactivated vaccines.
Collapse
|
33
|
Gasper DJ, Neldner B, Plisch EH, Rustom H, Carrow E, Imai H, Kawaoka Y, Suresh M. Effective Respiratory CD8 T-Cell Immunity to Influenza Virus Induced by Intranasal Carbomer-Lecithin-Adjuvanted Non-replicating Vaccines. PLoS Pathog 2016; 12:e1006064. [PMID: 27997610 PMCID: PMC5173246 DOI: 10.1371/journal.ppat.1006064] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 11/14/2016] [Indexed: 01/31/2023] Open
Abstract
CD8+ cytotoxic T lymphocytes (CTLs) are critical for clearing many viral infections, and protective CTL memory can be induced by vaccination with attenuated viruses and vectors. Non-replicating vaccines are typically potentiated by the addition of adjuvants that enhance humoral responses, however few are capable of generating CTL responses. Adjuplex is a carbomer-lecithin-based adjuvant demonstrated to elicit robust humoral immunity to non-replicating antigens. We report that mice immunized with non-replicating Adjuplex-adjuvanted vaccines generated robust antigen-specific CTL responses. Vaccination by the subcutaneous or the intranasal route stimulated systemic and mucosal CTL memory respectively. However, only CTL memory induced by intranasal vaccination was protective against influenza viral challenge, and correlated with an enhancement of memory CTLs in the airways and CD103+ CD69+ CXCR3+ resident memory-like CTLs in the lungs. Mechanistically, Myd88-deficient mice mounted primary CTL responses to Adjuplex vaccines that were similar in magnitude to wild-type mice, but exhibited altered differentiation of effector cell subsets. Immune potentiating effects of Adjuplex entailed alterations in the frequency of antigen-presenting-cell subsets in vaccine draining lymph nodes, and in the lungs and airways following intranasal vaccination. Further, Adjuplex enhanced the ability of dendritic cells to promote antigen-induced proliferation of naïve CD8 T cells by modulating antigen uptake, its intracellular localization, and rate of processing. Taken together, we have identified an adjuvant that elicits both systemic and mucosal CTL memory to non-replicating antigens, and engenders protective CTL-based heterosubtypic immunity to influenza A virus in the respiratory tract. Further, findings presented in this manuscript have provided key insights into the mechanisms and factors that govern the induction and programming of systemic and protective memory CTLs in the respiratory tract. Current respiratory-virus vaccines typically employ non-replicating antigens and rely solely on the generation of humoral responses for protection. Viruses such as influenza can mutate and escape these responses, thereby limiting immunity and necessitating revaccination. Cell-mediated immunity (CMI) could provide broader protection by targeting viral components that infrequently mutate, however non-replicating vaccines capable of inducing CMI are not available. Impediments to vaccine development include an incomplete understanding of the nature of protective respiratory CMI and a lack of vaccine adjuvants capable of eliciting CMI to non-replicating antigens. Using a mouse model, we characterized the protective immunity afforded by CMI responses to non-replicating vaccines formulated with the adjuvant Adjuplex. We found that vaccination via either the subcutaneous or intranasal route was capable of inducing potent CMI responses. However, only intranasal vaccination protected against challenge with heterosubtypic influenza viruses. This protection correlated with enhancement of T cells with a resident-memory phenotype in the lungs. Additionally, mechanistic studies showed that Adjuplex affects antigen-presenting cells via activation and alteration of antigen uptake, processing, and presentation. The current studies: (1) identified an adjuvant that elicits protective CMI to respiratory viral pathogens; (2) suggested that stimulation of protective CMI in the respiratory tract requires intranasal vaccine delivery.
Collapse
Affiliation(s)
- David J Gasper
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America.,Comparative Biomedical Sciences Graduate Program, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Brandon Neldner
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Erin H Plisch
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Hani Rustom
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Emily Carrow
- Advanced Bioadjuvants, Omaha, Nebraska, United States of America
| | - Hirotaka Imai
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - M Suresh
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
34
|
Size distribution analysis of influenza virus particles using size exclusion chromatography. J Chromatogr A 2016; 1465:117-25. [DOI: 10.1016/j.chroma.2016.08.056] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/23/2016] [Accepted: 08/25/2016] [Indexed: 01/17/2023]
|
35
|
Visualization and Sequencing of Membrane Remodeling Leading to Influenza Virus Fusion. J Virol 2016; 90:6948-6962. [PMID: 27226364 DOI: 10.1128/jvi.00240-16] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 05/14/2016] [Indexed: 12/31/2022] Open
Abstract
UNLABELLED Protein-mediated membrane fusion is an essential step in many fundamental biological events, including enveloped virus infection. The nature of protein and membrane intermediates and the sequence of membrane remodeling during these essential processes remain poorly understood. Here we used cryo-electron tomography (cryo-ET) to image the interplay between influenza virus and vesicles with a range of lipid compositions. By following the population kinetics of membrane fusion intermediates imaged by cryo-ET, we found that membrane remodeling commenced with the hemagglutinin fusion protein spikes grappling onto the target membrane, followed by localized target membrane dimpling as local clusters of hemagglutinin started to undergo conformational refolding. The local dimples then transitioned to extended, tightly apposed contact zones where the two proximal membrane leaflets were in most cases indistinguishable from each other, suggesting significant dehydration and possible intermingling of the lipid head groups. Increasing the content of fusion-enhancing cholesterol or bis-monoacylglycerophosphate in the target membrane led to an increase in extended contact zone formation. Interestingly, hemifused intermediates were found to be extremely rare in the influenza virus fusion system studied here, most likely reflecting the instability of this state and its rapid conversion to postfusion complexes, which increased in population over time. By tracking the populations of fusion complexes over time, the architecture and sequence of membrane reorganization leading to efficient enveloped virus fusion were thus resolved. IMPORTANCE Enveloped viruses employ specialized surface proteins to mediate fusion of cellular and viral membranes that results in the formation of pores through which the viral genetic material is delivered to the cell. For influenza virus, the trimeric hemagglutinin (HA) glycoprotein spike mediates host cell attachment and membrane fusion. While structures of a subset of conformations and parts of the fusion machinery have been characterized, the nature and sequence of membrane deformations during fusion have largely eluded characterization. Building upon studies that focused on early stages of HA-mediated membrane remodeling, here cryo-electron tomography (cryo-ET) was used to image the three-dimensional organization of intact influenza virions at different stages of fusion with liposomes, leading all the way to completion of the fusion reaction. By monitoring the evolution of fusion intermediate populations over the course of acid-induced fusion, we identified the progression of membrane reorganization that leads to efficient fusion by an enveloped virus.
Collapse
|
36
|
Analysis of the beta-propiolactone sensitivity and optimization of inactivation methods for human influenza H3N2 virus. J Virol Methods 2016; 235:105-111. [PMID: 27142111 DOI: 10.1016/j.jviromet.2016.04.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 04/21/2016] [Accepted: 04/21/2016] [Indexed: 11/21/2022]
Abstract
Beta-propiolactone (BPL) is used as an inactivating reagent for influenza virus in a number of countries. However, the treatment of viruses with BPL occasionally results in a decrease in the hemagglutinin (HA) titer, which complicates vaccine development. In the present study, we examined the biological and biochemical characteristics of human H1N1 and H3N2 viruses treated with BPL, and developed an inactivation method for BPL-sensitive viruses. A significant decrease in HA titer was detected in the H3N2 viruses examined. The decrease in the pH of the virus fluid was not associated with the decreased HA titer, indicating that the decrease in HA titer for the H3N2 virus is the result of the direct effect of BPL. Excessive modification of M1 by BPL and loss of virion diameter were observed in 0.1% BPL-treated H3N2 virus. Taken together, these results suggest that the BPL sensitivity of H3N2 virus results from disruption of the virion. By contrast, the H3N2 virus was successfully inactivated by 0.02% BPL without a significant decrease in the HA titer or disruption of virion structure. Furthermore, we found that the 0.02% BPL in the virion preparation was hydrolyzed successfully by incubation at 37°C for 7h. Thus, mild treatment with a low concentration of BPL enabled us to inactivate the H3N2 virus.
Collapse
|
37
|
The hemifusion structure induced by influenza virus haemagglutinin is determined by physical properties of the target membranes. Nat Microbiol 2016; 1:16050. [PMID: 27572837 PMCID: PMC4876733 DOI: 10.1038/nmicrobiol.2016.50] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 03/14/2016] [Indexed: 11/20/2022]
Abstract
Influenza A virus hemagglutinin (HA) changes conformation and drives membrane fusion of viral and endosomal membrane at low pH. Membrane fusion proceeds through an intermediate called hemifusion1,2. For viral fusion the hemifusion structures are not determined3. Here, influenza virus-like particles (VLP)4 carrying wild-type (WT) HA or HA hemifusion mutant G1S5 and liposome mixtures were studied at low pH by Volta phase plate (VPP) cryo-electron tomography (cET) which improves signal-to-noise ratio close to focus. We determined two distinct hemifusion structures: a hemifusion diaphragm (HD) and a novel structure termed lipidic junction. Liposomes with lipidic junctions were ruptured with membrane edges stabilized by HA. The rupture frequency and HD diameter were not affected by G1S mutation, but decreased when the cholesterol level in the liposomes was close to physiological concentrations. We propose that HA induces merger between the viral and a target membrane by one of two independent pathways: rupture-insertion pathway leading to lipidic junction and hemifusion-stalk pathway leading to fusion pore. The latter is relevant under the conditions of influenza virus infection of cells. Cholesterol concentration functions as a pathway switch due to its negative spontaneous curvature in the target bilayer as determined by continuum analysis.
Collapse
|
38
|
Study of rabies virus by Differential Scanning Calorimetry. Biochem Biophys Rep 2015; 4:329-336. [PMID: 29124221 PMCID: PMC5669403 DOI: 10.1016/j.bbrep.2015.10.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 10/22/2015] [Accepted: 10/23/2015] [Indexed: 12/25/2022] Open
Abstract
Differential Scanning Calorimetry (DSC) has been used in the past to study the thermal unfolding of many different viruses. Here we present the first DSC analysis of rabies virus. We show that non-inactivated, purified rabies virus unfolds cooperatively in two events centered at approximately 62 and 73 °C. Beta-propiolactone (BPL) treatment does not alter significantly viral unfolding behavior, indicating that viral inactivation does not alter protein structure significantly. The first unfolding event was absent in bromelain treated samples, causing an elimination of the G-protein ectodomain, suggesting that this event corresponds to G-protein unfolding. This hypothesis was confirmed by the observation that this first event was shifted to higher temperatures in the presence of three monoclonal, G-protein specific antibodies. We show that dithiothreitol treatment of the virus abolishes the first unfolding event, indicating that the reduction of G-protein disulfide bonds causes dramatic alterations to protein structure. Inactivated virus samples heated up to 70 °C also showed abolished recognition of conformational G-protein specific antibodies by Surface Plasmon Resonance analysis. The sharpness of unfolding transitions and the low standard deviations of the Tm values as derived from multiple analysis offers the possibility of using this analytical tool for efficient monitoring of the vaccine production process and lot to lot consistency. Differential Scanning Calorimetry analysis of rabies virus. Rabies virus unfolds in two thermal events. The first event corresponds to G-protein.
Collapse
|
39
|
Fontana J, Steven AC. Influenza virus-mediated membrane fusion: Structural insights from electron microscopy. Arch Biochem Biophys 2015; 581:86-97. [PMID: 25958107 DOI: 10.1016/j.abb.2015.04.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 04/21/2015] [Accepted: 04/27/2015] [Indexed: 12/26/2022]
Abstract
Influenza virus, the causative agent of flu, enters the host cell by endocytosis. The low pH encountered inside endosomes triggers conformational changes in the viral glycoprotein hemagglutinin (HA), that mediate fusion of the viral and cellular membranes. This releases the viral genome into the cytoplasm of the infected cell, establishing the onset of the replication cycle. To investigate the structural basis of HA-mediated membrane fusion, a number of techniques have been employed. These include X-ray crystallography, which has provided atomic models of the HA ectodomain in its initial (pre-fusion) state and of part of HA in its final (post-fusion) state. However, this left an information deficit concerning many other aspects of the fusion process. Electron microscopy (EM) approaches are helping to fill this void. For example, influenza virions at neutral pH have been imaged by cryo-EM and cryo-electron tomography (cryo-ET); thin section EM has shown that influenza viruses enter the cell by endocytosis; the large-scale structural changes in HA when virions are exposed to low pH (pre-fusion to post-fusion states) have been visualized by negative staining and cryo-EM; acidification also induces structural changes in the M1 matrix layer and its separation from the viral envelope; intermediate HA conformations between its pre- and post-fusion states have been detected by cryo-ET supplemented with subtomogram averaging; and fusion of influenza virions with liposomes has been visualized by cryo-ET. In this review, we survey EM-based contributions towards the characterization of influenza virus-mediated membrane fusion and anticipate the potential for future developments.
Collapse
Affiliation(s)
- Juan Fontana
- Laboratory of Structural Biology Research, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Alasdair C Steven
- Laboratory of Structural Biology Research, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
40
|
Godefroy C, Dahmane S, Dosset P, Adam O, Nicolai MC, Ronzon F, Milhiet PE. Mimicking influenza virus fusion using supported lipid bilayers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2014; 30:11394-11400. [PMID: 25186242 DOI: 10.1021/la502591a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Influenza virus infection is a serious public health problem in the world, and understanding the molecular mechanisms involved in viral replication is crucial. In this paper, we used a minimalist approach based on a lipid bilayer supported on mica, which we imaged by atomic force microscopy (AFM) in a physiological buffer, to analyze the different steps of influenza fusion, from the interaction of intact viruses with the supported bilayer to their complete fusion. Our results show that sialic acid recognition and priming upon acidification are sufficient for a complete fusion with the host cell membrane. After fusion, a flat and continuous membrane was observed. Because of the fragility of the viral membrane that was removed by the tip, most probably due to the disorganization of the matrix layer at acidic pH, fine structural details of ribonucleoproteins (RNP) were obtained. In addition, AFM topography of intact virus in interaction with the supported lipid bilayer confirms that hemeagglutinin and neuraminidase can form isolated clusters within the viral membrane.
Collapse
Affiliation(s)
- Cédric Godefroy
- Institut National de la Santé et de la Recherche Médicale, Unité 1054, 34090 Montpellier, France
| | | | | | | | | | | | | |
Collapse
|
41
|
Stepwise priming by acidic pH and a high K+ concentration is required for efficient uncoating of influenza A virus cores after penetration. J Virol 2014; 88:13029-46. [PMID: 25165113 DOI: 10.1128/jvi.01430-14] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
UNLABELLED Influenza A virus (IAV) uses the low pH in late endocytic vacuoles as a cue for penetration by membrane fusion. Here, we analyzed the prefusion reactions that prepare the core for uncoating after it has been delivered to the cytosol. We found that this priming process occurs in two steps that are mediated by the envelope-embedded M2 ion channel. The first weakens the interactions between the matrix protein, M1, and the viral ribonucleoprotein bundle. It involves a conformational change in a linker sequence and the C-terminal domain of M1 after exposure to a pH below 6.5. The second step is triggered by a pH of <6.0 and by the influx of K(+) ions. It causes additional changes in M1 as well as a loss of stability in the viral ribonucleoprotein bundle. Our results indicate that both the switch from Na(+) to K(+) in maturing endosomes and the decreasing pH are needed to prime IAV cores for efficient uncoating and infection of the host cell. IMPORTANCE The entry of IAV involves several steps, including endocytosis and fusion at late endosomes. Entry also includes disassembly of the viral core, which is composed of the viral ribonucleoproteins and the RNA genome. We have found that the uncoating process of IAV is initiated long before the core is delivered into the cytosol. M2, an ion channel in the viral membrane, is activated when the virus passes through early endosomes. Here, we show that protons entering the virus through M2 cause a conformational change in the matrix protein, M1. This weakens interactions between M1 and the viral ribonucleoproteins. A second change was found to occur when the virus enters late endosomes. The preacidified core is then exposed to a high concentration of K(+), which affects the interactions between the ribonucleoproteins. Thus, when cores are finally delivered to the cytosol, they are already partially destabilized and, therefore, uncoating competent and infectious.
Collapse
|
42
|
Development of an algorithm for production of inactivated arbovirus antigens in cell culture. J Virol Methods 2014; 208:66-78. [PMID: 25102428 DOI: 10.1016/j.jviromet.2014.07.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 07/17/2014] [Accepted: 07/26/2014] [Indexed: 11/22/2022]
Abstract
Arboviruses are medically important pathogens that cause human disease ranging from a mild fever to encephalitis. Laboratory diagnosis is essential to differentiate arbovirus infections from other pathogens with similar clinical manifestations. The Arboviral Diseases Branch (ADB) reference laboratory at the CDC Division of Vector-Borne Diseases (DVBD) produces reference antigens used in serological assays such as the virus-specific immunoglobulin M antibody-capture enzyme-linked immunosorbent assay (MAC-ELISA). Antigen production in cell culture has largely replaced the use of suckling mice; however, the methods are not directly transferable. The development of a cell culture antigen production algorithm for nine arboviruses from the three main arbovirus families, Flaviviridae, Togaviridae, and Bunyaviridae, is described here. Virus cell culture growth and harvest conditions were optimized, inactivation methods were evaluated, and concentration procedures were compared for each virus. Antigen performance was evaluated by the MAC-ELISA at each step of the procedure. The antigen production algorithm is a framework for standardization of methodology and quality control; however, a single antigen production protocol was not applicable to all arboviruses and needed to be optimized for each virus.
Collapse
|