65 results on '"Wirblich C"'
Search Results
2. Expression and Functional Characterization of Bluetongue Virus VP5 Protein: Role in Cellular Permeabilization
- Author
-
Hassan, S. H., primary, Wirblich, C., additional, Forzan, M., additional, and Roy, P., additional
- Published
- 2001
- Full Text
- View/download PDF
3. Genetic map of the calicivirus rabbit hemorrhagic disease virus as deduced from in vitro translation studies
- Author
-
Wirblich, C, primary, Thiel, H J, additional, and Meyers, G, additional
- Published
- 1996
- Full Text
- View/download PDF
4. 3C-like protease of rabbit hemorrhagic disease virus: identification of cleavage sites in the ORF1 polyprotein and analysis of cleavage specificity
- Author
-
Wirblich, C, primary, Sibilia, M, additional, Boniotti, M B, additional, Rossi, C, additional, Thiel, H J, additional, and Meyers, G, additional
- Published
- 1995
- Full Text
- View/download PDF
5. Identification and characterization of a 3C-like protease from rabbit hemorrhagic disease virus, a calicivirus
- Author
-
Boniotti, B, primary, Wirblich, C, additional, Sibilia, M, additional, Meyers, G, additional, Thiel, H J, additional, and Rossi, C, additional
- Published
- 1994
- Full Text
- View/download PDF
6. European brown hare syndrome virus: relationship to rabbit hemorrhagic disease virus and other caliciviruses
- Author
-
Wirblich, C, primary, Meyers, G, additional, Ohlinger, V F, additional, Capucci, L, additional, Eskens, U, additional, Haas, B, additional, and Thiel, H J, additional
- Published
- 1994
- Full Text
- View/download PDF
7. Toward the Development of a Pan-Lyssavirus Vaccine.
- Author
-
Ben Hamed S, Myers JF, Chandwani A, Wirblich C, Kurup D, Paran N, and Schnell MJ
- Subjects
- Animals, Mice, Rhabdoviridae Infections prevention & control, Rhabdoviridae Infections immunology, Rhabdoviridae Infections veterinary, Rhabdoviridae Infections virology, Vaccines, Synthetic immunology, Vaccines, Synthetic administration & dosage, Female, Viral Vaccines immunology, Glycoproteins immunology, Glycoproteins genetics, Antibodies, Neutralizing immunology, Antibodies, Neutralizing blood, Vaccine Development, Humans, Antigens, Viral immunology, Mice, Inbred BALB C, Lyssavirus immunology, Lyssavirus genetics, Antibodies, Viral immunology, Antibodies, Viral blood, Rabies virus immunology, Rabies virus genetics, Rabies Vaccines immunology, Rabies Vaccines administration & dosage, Rabies prevention & control, Rabies immunology, Rabies virology
- Abstract
In addition to the rabies virus (RABV), 16 more lyssavirus species have been identified worldwide, causing a disease similar to RABV. Non-rabies-related human deaths have been described, but the number of cases is unknown, and the potential of such lyssaviruses causing human disease is unpredictable. The current rabies vaccine does not protect against divergent lyssaviruses such as Mokola virus (MOKV) or Lagos bat virus (LBV). Thus, a more broad pan-lyssavirus vaccine is needed. Here, we evaluate a novel lyssavirus vaccine with an attenuated RABV vector harboring a chimeric RABV glycoprotein (G) in which the antigenic site I of MOKV replaces the authentic site of rabies virus (RABVG-cAS1). The recombinant vaccine was utilized to immunize mice and analyze the immune response compared to homologous vaccines. Our findings indicate that the vaccine RABVG-cAS1 was immunogenic and induced high antibody titers against both RABVG and MOKVG. Challenge studies with different lyssaviruses showed that replacing a single antigenic site of RABV G with the corresponding site of MOKV G provides a significant improvement over the homologous RABV vaccine and protects against RABV, Irkut virus (IRKV), and MOKV. This strategy of epitope chimerization paves the way towards a pan-lyssavirus vaccine to safely combat the diseases caused by these viruses.
- Published
- 2024
- Full Text
- View/download PDF
8. Effects of adjuvants in a rabies-vectored Ebola virus vaccine on protection from surrogate challenge.
- Author
-
Yankowski C, Kurup D, Wirblich C, and Schnell MJ
- Abstract
Ebola virus is the primary contributor to the global threat of filovirus severe hemorrhagic fever, and Ebola virus disease has a case fatality rate of 50-90%. An inactivated, bivalent filovirus/rabies virus vaccine, FILORAB1, consists of recombinant rabies virus virions expressing the Ebola virus glycoprotein. FILORAB1 is immunogenic and protective from Ebola virus challenge in mice and non-human primates, and protection is enhanced when formulated with toll-like receptor 4 agonist Glucopyranosyl lipid adjuvant (GLA) in a squalene oil-in-water emulsion (SE). Through an adjuvant comparison in mice, we demonstrate that GLA-SE improves FILORAB1 efficacy by activating the innate immune system and shaping a Th1-biased adaptive immune response. GLA-SE adjuvanted mice and those adjuvanted with the SE component are better protected from surrogate challenge, while Th2 alum adjuvanted mice are not. Additionally, the immune response to FILORAB1 is long-lasting, as exhibited by highly-maintained serum antibody titers and long-lived cells in the spleen and bone marrow., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
9. Inactivated rabies-vectored SARS-CoV-2 vaccine provides long-term immune response unaffected by vector immunity.
- Author
-
Yankowski C, Wirblich C, Kurup D, and Schnell MJ
- Abstract
The objective of this study is to further analyze recombinant rabies virus-vectored SARS-CoV-2 vaccine, CORAVAX, as an effective COVID-19 vaccine strategy. CORAVAX has proven immunogenic and protective against SARS-CoV-2 in animal models. Here, we have screened adjuvants for the highest quality antibody titers, negated the concern of pre-existing rabies-vector immunity, and established its potential as a long-term COVID-19 vaccine. We have tested toll-like receptor 4 (TLR4) agonists, inflammasome activators, and alum adjuvants in CORAVAX and found TLR4-activating MPLA-AddaVax to have the greatest potential. We followed the humoral immune response to CORAVAX in mice with pre-existing rabies virus immunity and saw no significant differences compared to naive mice. We then followed the immune response to CORAVAX over several months and 1-year post-immunization. Mice maintained high antigen-specific serum antibody titers as well as long-lived antibody-secreting cells in the spleen and bone marrow. We believe this rabies-vector strategy combats the problem of waning immunity of other COVID-19 vaccines. These results together support CORAVAX's potential during the ongoing COVID-19 pandemic., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
10. A Single Dose of the Deactivated Rabies-Virus Vectored COVID-19 Vaccine, CORAVAX, Is Highly Efficacious and Alleviates Lung Inflammation in the Hamster Model.
- Author
-
Kurup D, Wirblich C, Zabihi Diba L, Lambert R, Watson M, Shaikh N, Ramage H, Solomides C, and Schnell MJ
- Subjects
- Animals, Antibodies, Neutralizing, Antibodies, Viral, COVID-19 Vaccines, Cricetinae, Humans, SARS-CoV-2 genetics, Spike Glycoprotein, Coronavirus, COVID-19 prevention & control, Rabies prevention & control, Rabies Vaccines, Rabies virus, Viral Vaccines
- Abstract
Without sufficient herd immunity through either vaccination or natural infection, the coronavirus disease 2019 pandemic is unlikely to be controlled. Waning immunity with the currently approved vaccines suggests the need to evaluate vaccines causing the induction of long-term responses. Here, we report the immunogenicity and efficacy of our adjuvanted single-dose Rabies-vectored SARS-CoV-2 S1 vaccine, CORAVAX, in hamsters. CORAVAX induces high SARS-CoV-2 S1-specific and virus-neutralizing antibodies (VNAs) that prevent weight loss, viral loads, disease, lung inflammation, and the cytokine storm in hamsters. We also observed high Rabies VNA titers. In summary, CORAVAX is a promising dual-antigen vaccine candidate for clinical evaluation against SARS-CoV-2 and Rabies virus.
- Published
- 2022
- Full Text
- View/download PDF
11. Measles-based Zika vaccine induces long-term immunity and requires NS1 antibodies to protect the female reproductive tract.
- Author
-
Kurup D, Wirblich C, Lambert R, Diba LZ, Leiby BE, and Schnell MJ
- Abstract
Zika virus (ZIKV) can cause devastating effects in the unborn fetus of pregnant women. To develop a candidate vaccine that can protect human fetuses, we generated a panel of live measles vaccine (MV) vectors expressing ZIKV-E and -NS1. Our MV-based ZIKV-E vaccine, MV-E2, protected mice from the non-lethal Zika Asian strain (PRVABC59) and the lethal African strain (MR766) challenge. Despite 100% survival of the MV-E2 mice, however, complete viral clearance was not achieved in the brain and reproductive tract of the lethally challenged mice. We then tested MV-based vaccines that expressed E and NS1 together or separately in two different vaccines. We observed complete clearance of ZIKV from the female reproductive tract and complete fetal protection in the lethal African challenge model in animals that received the dual antigen vaccines. Additionally, MV-E2 and MV-NS1, when administered together, induced durable plasma cell responses. Our findings suggest that NS1 antibodies are required to enhance the protection of ZIKV-E antibodies in the female reproductive tract., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
12. A Strategy to Detect Emerging Non-Delta SARS-CoV-2 Variants with a Monoclonal Antibody Specific for the N501 Spike Residue.
- Author
-
Puligedda RD, Al-Saleem FH, Wirblich C, Kattala CD, Jović M, Geiszler L, Devabhaktuni H, Feuerstein GZ, Schnell MJ, Sack M, Livornese LL Jr, and Dessain SK
- Abstract
Efforts to control SARS-CoV-2 have been challenged by the emergence of variant strains that have important implications for clinical and epidemiological decision making. Four variants of concern (VOCs) have been designated by the Centers for Disease Control and Prevention (CDC), namely, B.1.617.2 (delta), B.1.1.7 (alpha), B.1.351 (beta), and P.1 (gamma), although the last three have been downgraded to variants being monitored (VBMs). VOCs and VBMs have shown increased transmissibility and/or disease severity, resistance to convalescent SARS-CoV-2 immunity and antibody therapeutics, and the potential to evade diagnostic detection. Methods are needed for point-of-care (POC) testing to rapidly identify these variants, protect vulnerable populations, and improve surveillance. Antigen-detection rapid diagnostic tests (Ag-RDTs) are ideal for POC use, but Ag-RDTs that recognize specific variants have not yet been implemented. Here, we describe a mAb (2E8) that is specific for the SARS-CoV-2 spike protein N501 residue. The 2E8 mAb can distinguish the delta VOC from variants with the N501Y meta-signature, which is characterized by convergent mutations that contribute to increased virulence and evasion of host immunity. Among the N501Y-containing mutants formerly designated as VOCs (alpha, beta, and gamma), a previously described mAb, CB6, can distinguish beta from alpha and gamma. When used in a sandwich ELISA, these mAbs sort these important SARS-CoV-2 variants into three diagnostic categories, namely, (1) delta, (2) alpha or gamma, and (3) beta. As delta is currently the predominant variant globally, they will be useful for POC testing to identify N501Y meta-signature variants, protect individuals in high-risk settings, and help detect epidemiological shifts among SARS-CoV-2 variants.
- Published
- 2021
- Full Text
- View/download PDF
13. Tetravalent Rabies-Vectored Filovirus and Lassa Fever Vaccine Induces Long-term Immunity in Nonhuman Primates.
- Author
-
Kurup D, Fisher CR, Scher G, Yankowski C, Testa A, Keshwara R, Abreu-Mota T, Lambert R, Ferguson M, Rinaldi W, Ruiz L, Wirblich C, and Schnell MJ
- Subjects
- Animals, Antibodies, Viral blood, Macaca fascicularis, Marburgvirus immunology, Vaccines, Combined, Ebolavirus immunology, Hemorrhagic Fever, Ebola prevention & control, Lassa Fever prevention & control, Lassa virus immunology, Rabies prevention & control, Rabies Vaccines administration & dosage
- Abstract
Background: The objective of this study is to evaluate the immunogenicity of adjuvanted monovalent rabies virus (RABV)-based vaccine candidates against Ebola virus (FILORAB1), Sudan virus (FILORAB2), Marburg virus (FILORAB3), Lassa virus (LASSARAB1), and combined trivalent vaccine candidate (FILORAB1-3) and tetravalent vaccine candidate (FILORAB1-3 and LASSARAB) in nonhuman primates., Methods: Twenty-four Macaca fascicularis were randomly assigned into 6 groups of 4 animals. Each group was vaccinated with either a single adjuvanted vaccine, the trivalent vaccine, or the tetravalent vaccine at days 0 and 28. We followed the humoral immune responses for 1 year by antigen-specific enzyme-linked immunosorbent assays and RABV neutralization assays., Results: High titers of filovirus and/or Lassa virus glycoprotein-specific immunoglobulin G were induced in the vaccinated animals. There were no significant differences between immune responses in animals vaccinated with single vaccines vs trivalent or tetravalent vaccines. In addition, all vaccine groups elicited strong rabies neutralizing antibody titers. The antigen-specific immune responses were detectable for 1 year in all groups., Conclusions: In summary, this study shows the longevity of the immune responses up to 365 days for a pentavalent vaccine-against Ebola virus, Sudan virus, Marburg virus, Lassa virus, and RABV-using a safe and effective vaccine platform., (© The Author(s) 2021. Published by Oxford University Press for the Infectious Diseases Society of America. All rights reserved. For permissions, e-mail: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
14. A single dose of replication-competent VSV-vectored vaccine expressing SARS-CoV-2 S1 protects against virus replication in a hamster model of severe COVID-19.
- Author
-
Malherbe DC, Kurup D, Wirblich C, Ronk AJ, Mire C, Kuzmina N, Shaik N, Periasamy S, Hyde MA, Williams JM, Shi PY, Schnell MJ, and Bukreyev A
- Abstract
The development of effective countermeasures against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the agent responsible for the COVID-19 pandemic, is a priority. We designed and produced ConVac, a replication-competent vesicular stomatitis virus (VSV) vaccine vector that expresses the S1 subunit of SARS-CoV-2 spike protein. We used golden Syrian hamsters as animal models of severe COVID-19 to test the efficacy of the ConVac vaccine. A single vaccine dose elicited high levels of SARS-CoV-2 specific binding and neutralizing antibodies; following intranasal challenge with SARS-CoV-2, animals were protected from weight loss and viral replication in the lungs. No enhanced pathology was observed in vaccinated animals upon challenge, but some inflammation was still detected. The data indicate rapid control of SARS-CoV-2 replication by the S1-based VSV-vectored SARS-CoV-2 ConVac vaccine., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
15. Inactivated rabies virus vectored SARS-CoV-2 vaccine prevents disease in a Syrian hamster model.
- Author
-
Kurup D, Malherbe DC, Wirblich C, Lambert R, Ronk AJ, Zabihi Diba L, Bukreyev A, and Schnell MJ
- Subjects
- Animals, Disease Models, Animal, Humans, Mesocricetus, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, COVID-19 immunology, COVID-19 prevention & control, COVID-19 Vaccines immunology, Rabies virus immunology, SARS-CoV-2 immunology
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an emergent coronavirus that has caused a worldwide pandemic. Although human disease is often asymptomatic, some develop severe illnesses such as pneumonia, respiratory failure, and death. There is an urgent need for a vaccine to prevent its rapid spread as asymptomatic infections accounting for up to 40% of transmission events. Here we further evaluated an inactivated rabies vectored SARS-CoV-2 S1 vaccine CORAVAX in a Syrian hamster model. CORAVAX adjuvanted with MPLA-AddaVax, a TRL4 agonist, induced high levels of neutralizing antibodies and generated a strong Th1-biased immune response. Vaccinated hamsters were protected from weight loss and viral replication in the lungs and nasal turbinates three days after challenge with SARS-CoV-2. CORAVAX also prevented lung disease, as indicated by the significant reduction in lung pathology. This study highlights CORAVAX as a safe, immunogenic, and efficacious vaccine that warrants further assessment in human trials., Competing Interests: I have read the journal’s policy and the authors of this manuscript have the following competing interests: “M.J.S., C.W., and D.K. are coinventors of the patent application “Coronavirus disease (COVID-19) vaccine”. A.B, D.C.M, A.J.R., R.L., and L.Z.B. have no competing interests.”
- Published
- 2021
- Full Text
- View/download PDF
16. Rabies virus-based COVID-19 vaccine CORAVAX™ induces high levels of neutralizing antibodies against SARS-CoV-2.
- Author
-
Kurup D, Wirblich C, Ramage H, and Schnell MJ
- Abstract
The recently emerged coronavirus SARS-CoV-2, the causative agent of COVID-19, is rapidly spreading in the world. The exponentially expanding threat of SARS-CoV-2 to global health highlights the urgent need for a vaccine. Herein we show the rapid development of a novel, highly efficient, and safe COVID-19 vaccine using a rabies virus-based vector that has proven to be an efficient vaccine against several emerging infectious diseases. This study reports that both a live and an inactivated rabies virus containing the SARS-CoV-2 spike S1 protein induces potent virus-neutralizing antibodies at much higher levels than seen in the sera of convalescent patients. In summary, the results provided here warrant further development of this safe and established vaccine platform against COVID-19., Competing Interests: Competing interestsM.J.S., C.W. and D.K. are coinventors of the patent application “Coronavirus disease (COVID-19) vaccine”. H.R. declares that there are no competing interests., (© The Author(s) 2020.)
- Published
- 2020
- Full Text
- View/download PDF
17. Lyssavirus Vaccine with a Chimeric Glycoprotein Protects across Phylogroups.
- Author
-
Fisher CR, Lowe DE, Smith TG, Yang Y, Hutson CL, Wirblich C, Cingolani G, and Schnell MJ
- Subjects
- Administration, Intranasal, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Glycoproteins chemistry, Immunity, Humoral, Injections, Intramuscular, Rabies Vaccines immunology, Recombinant Proteins chemistry, Virus Replication physiology, Glycoproteins metabolism, Lyssavirus immunology, Phylogeny, Recombinant Proteins metabolism, Viral Vaccines immunology
- Abstract
Rabies is nearly 100% lethal in the absence of treatment, killing an estimated 59,000 people annually. Vaccines and biologics are highly efficacious when administered properly. Sixteen rabies-related viruses (lyssaviruses) are similarly lethal, but some are divergent enough to evade protection from current vaccines and biologics, which are based only on the classical rabies virus (RABV). Here we present the development and characterization of LyssaVax, a vaccine featuring a structurally designed, functional chimeric glycoprotein (G) containing immunologically important domains from both RABV G and the highly divergent Mokola virus (MOKV) G. LyssaVax elicits high titers of antibodies specific to both RABV and MOKV Gs in mice. Immune sera also neutralize a range of wild-type lyssaviruses across the major phylogroups. LyssaVax-immunized mice are protected against challenge with recombinant RABV and MOKV. Altogether, LyssaVax demonstrates the utility of structural modeling in vaccine design and constitutes a broadened lyssavirus vaccine candidate., Competing Interests: Declaration of Interests C.R.F., C.W., and M.J.S. are inventors on the U.S. Patent Application No. WO2018231974A1 (“Composition and Administration of Chimeric Glycoprotein Lyssavirus Vaccines for Coverage against Rabies”)., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
18. Identification and Characterization of a Small-Molecule Rabies Virus Entry Inhibitor.
- Author
-
Du Pont V, Wirblich C, Yoon JJ, Cox RM, Schnell MJ, and Plemper RK
- Subjects
- Animals, Antibodies, Viral immunology, Antiviral Agents pharmacology, Cell Line, Cross Protection, Humans, Peptide Library, Rabies prevention & control, Rabies Vaccines immunology, Rabies virus metabolism, Rabies virus pathogenicity, Vesicular stomatitis Indiana virus genetics, Vesicular stomatitis Indiana virus immunology, Vesiculovirus genetics, Vesiculovirus immunology, Viral Fusion Proteins pharmacology, Antibodies, Neutralizing therapeutic use, Drug Evaluation, Preclinical methods, Rabies virus immunology
- Abstract
Rabies virus (RABV) causes a severe and fatal neurological disease, but morbidity is vaccine preventable and treatable prior to the onset of clinical symptoms. However, immunoglobulin (IgG)-based rabies postexposure prophylaxis (PEP) is expensive, restricting access to life-saving treatment, especially for patients in low-income countries where the clinical need is greatest, and does not confer cross-protection against newly emerging phylogroup II lyssaviruses. Toward identifying a cost-effective replacement for the IgG component of rabies PEP, we developed and implemented a high-throughput screening protocol utilizing a single-cycle RABV reporter strain. A large-scale screen and subsequent direct and orthogonal counterscreens identified a first-in-class direct-acting RABV inhibitor, GRP-60367, with a specificity index (SI) of >100,000. Mechanistic characterization through time-of-addition studies, transient cell-to-cell fusion assays, and chimeric vesicular stomatitis virus (VSV) recombinants expressing the RABV glycoprotein (G) demonstrated that GRP-60367 inhibits entry of a subset of RABV strains. Resistance profiling of the chemotype revealed hot spots in conserved hydrophobic positions of the RABV G protein fusion loop that were confirmed in transient cell-to-cell fusion assays. Transfer of RABV G genes with signature resistance mutations into a recombinant VSV backbone resulted in the recovery of replication-competent virions with low susceptibility to the inhibitor. This work outlines a tangible strategy for mechanistic characterization and resistance profiling of RABV drug candidates and identified a novel, well-behaved molecular probe chemotype that specifically targets the RABV G protein and prevents G-mediated viral entry. IMPORTANCE Rabies PEP depends on anti-RABV IgG, which is expensive and in limited supply in geographical areas with the highest disease burden. Replacing the IgG component with a cost-effective and shelf-stable small-molecule antiviral could address this unmet clinical need by expanding access to life-saving medication. This study has established a robust protocol for high-throughput anti-RABV drug screens and identified a chemically well-behaved, first-in-class hit with nanomolar anti-RABV potency that blocks RABV G protein-mediated viral entry. Resistance mapping revealed a druggable site formed by the G protein fusion loops that has not previously emerged as a target for neutralizing antibodies. Discovery of this RABV entry inhibitor establishes a new molecular probe to advance further mechanistic and structural characterization of RABV G that may aid in the design of a next-generation clinical candidate against RABV., (Copyright © 2020 American Society for Microbiology.)
- Published
- 2020
- Full Text
- View/download PDF
19. Erratum: Publisher Correction: Rabies-based vaccine induces potent immune responses against Nipah virus.
- Author
-
Keshwara R, Shiels T, Postnikova E, Kurup D, Wirblich C, Johnson RF, and Schnell MJ
- Abstract
[This corrects the article DOI: 10.1038/s41541-019-0109-5.].
- Published
- 2019
- Full Text
- View/download PDF
20. A new recombinant rabies virus expressing a green fluorescent protein: A novel and fast approach to quantify virus neutralizing antibodies.
- Author
-
Qin S, Volokhov D, Rodionova E, Wirblich C, Schnell MJ, Chizhikov V, and Dabrazhynetskaya A
- Subjects
- Animals, Antibodies, Neutralizing metabolism, Cell Line, Cell Line, Tumor, Fluorescence, Green Fluorescent Proteins chemistry, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Guinea Pigs, Humans, Luminescent Measurements methods, Mice, Neutralization Tests, Rabbits, Rabies prevention & control, Rabies virology, Rabies Vaccines administration & dosage, Rabies virus genetics, Rabies virus metabolism, Recombination, Genetic, Antibodies, Neutralizing immunology, Rabies immunology, Rabies Vaccines immunology, Rabies virus immunology
- Abstract
The Rapid Fluorescent Focus Inhibition Test (RFFIT) is a standard assay used to detect and assess the titers of rabies virus neutralizing antibodies (RVNA) in blood sera. To simplify the multistep RFFIT procedure by eliminating the immunostaining step, we generated a new recombinant RV expressing a green fluorescent protein (rRV-GFP) and assess its suitability for quantifying RVNA. We rescued the rRV-GFP virus from plasmid DNA carrying a full-length genome of the CVS-N2c strain of RV in which the eGFP gene was inserted between the glycoprotein and RNA-polymerase genes. The recombinant virus was genetically stable and grew efficiently in appropriate cells expressing sufficient GFP fluorescence to detect directly 20 h post infection (hpi). We evaluated the feasibility of using rRV-GFP in RFFIT by comparing RVNA titers in 27 serum samples measured by conventional RFFIT and RFFIT-GFP. A linear regression analysis of the data demonstrated a good agreement between these two methods (r = 0.9776) including results with samples having RVNA titers close to the minimally acceptable vaccine potency threshold (0.5 IU/ml). Study results showed that the rRV-GFP virus could replace the CVS-11 challenge virus currently used in the conventional RFFIT and enabling more rapid, simpler, and less expensive detection and quantitation of RVNA., (Copyright © 2019. Published by Elsevier Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
21. Rabies-based vaccine induces potent immune responses against Nipah virus.
- Author
-
Keshwara R, Shiels T, Postnikova E, Kurup D, Wirblich C, Johnson RF, and Schnell MJ
- 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., Competing Interests: The authors declare no competing interests.
- Published
- 2019
- Full Text
- View/download PDF
22. A Recombinant Rabies Virus Expressing the Marburg Virus Glycoprotein Is Dependent upon Antibody-Mediated Cellular Cytotoxicity for Protection against Marburg Virus Disease in a Murine Model.
- Author
-
Keshwara R, Hagen KR, Abreu-Mota T, Papaneri AB, Liu D, Wirblich C, Johnson RF, and Schnell MJ
- Subjects
- Animals, Cell Line, Chlorocebus aethiops, Disease Models, Animal, Female, HEK293 Cells, Humans, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Rabies Vaccines immunology, Vaccination methods, Vero Cells, Viral Vaccines immunology, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Glycoproteins immunology, Marburg Virus Disease immunology, Marburgvirus immunology, Rabies immunology, Rabies virus immunology
- Abstract
Marburg virus (MARV) is a filovirus related to Ebola virus (EBOV) associated with human hemorrhagic disease. Outbreaks are sporadic and severe, with a reported case mortality rate of upward of 88%. There is currently no antiviral or vaccine available. Given the sporadic nature of outbreaks, vaccines provide the best approach for long-term control of MARV in regions of endemicity. We have developed an inactivated rabies virus-vectored MARV vaccine (FILORAB3) to protect against Marburg virus disease. Immunogenicity studies in our labs have shown that a Th1-biased seroconversion to both rabies virus and MARV glycoproteins (GPs) is beneficial for protection in a preclinical murine model. As such, we adjuvanted FILORAB3 with glucopyranosyl lipid adjuvant (GLA), a Toll-like receptor 4 agonist, in a squalene-in-water emulsion. Across two different BALB/c mouse challenge models, we achieved 92% protection against murine-adapted Marburg virus (ma-MARV). Although our vaccine elicited strong MARV GP antibodies, it did not strongly induce neutralizing antibodies. Through both in vitro and in vivo approaches, we elucidated a critical role for NK cell-dependent antibody-mediated cellular cytotoxicity (ADCC) in vaccine-induced protection. Overall, these findings demonstrate that FILORAB3 is a promising vaccine candidate for Marburg virus disease. IMPORTANCE Marburg virus (MARV) is a virus similar to Ebola virus and also causes a hemorrhagic disease which is highly lethal. In contrast to EBOV, only a few vaccines have been developed against MARV, and researchers do not understand what kind of immune responses are required to protect from MARV. Here we show that antibodies directed against MARV after application of our vaccine protect in an animal system but fail to neutralize the virus in a widely used virus neutralization assay against MARV. This newly discovered activity needs to be considered more when analyzing MARV vaccines or infections., (Copyright © 2019 American Society for Microbiology.)
- Published
- 2019
- Full Text
- View/download PDF
23. Non-neutralizing antibodies elicited by recombinant Lassa-Rabies vaccine are critical for protection against Lassa fever.
- Author
-
Abreu-Mota T, Hagen KR, Cooper K, Jahrling PB, Tan G, Wirblich C, Johnson RF, and Schnell MJ
- Subjects
- 3T3 Cells, Animals, Antibody-Dependent Cell Cytotoxicity immunology, Cell Membrane metabolism, Genetic Vectors metabolism, Glucosides, Glycoproteins metabolism, Guinea Pigs, Immunity, Humoral, Immunization, Immunoglobulin G metabolism, Lassa Fever virology, Lassa virus pathogenicity, Lipid A, Macrophages metabolism, Mice, Mice, Inbred C57BL, Receptors, IgG metabolism, Virion metabolism, Virulence, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Lassa Fever immunology, Lassa Fever prevention & control, Lassa virus immunology, Rabies Vaccines immunology, Vaccines, Synthetic immunology
- Abstract
Lassa fever (LF), caused by Lassa virus (LASV), is a viral hemorrhagic fever for which no approved vaccine or potent antiviral treatment is available. LF is a WHO priority disease and, together with rabies, a major health burden in West Africa. Here we present the development and characterization of an inactivated recombinant LASV and rabies vaccine candidate (LASSARAB) that expresses a codon-optimized LASV glycoprotein (coGPC) and is adjuvanted by a TLR-4 agonist (GLA-SE). LASSARAB elicits lasting humoral response against LASV and RABV in both mouse and guinea pig models, and it protects both guinea pigs and mice against LF. We also demonstrate a previously unexplored role for non-neutralizing LASV GPC-specific antibodies as a major mechanism of protection by LASSARAB against LF through antibody-dependent cellular functions. Overall, these findings demonstrate an effective inactivated LF vaccine and elucidate a novel humoral correlate of protection for LF.
- Published
- 2018
- Full Text
- View/download PDF
24. Retrograde axonal transport of rabies virus is unaffected by interferon treatment but blocked by emetine locally in axons.
- Author
-
MacGibeny MA, Koyuncu OO, Wirblich C, Schnell MJ, and Enquist LW
- Subjects
- Animals, Emetine pharmacology, Interferons pharmacology, Protein Synthesis Inhibitors pharmacology, Rabies virus metabolism, Rats, Rats, Sprague-Dawley, Axonal Transport drug effects, Axons virology, Rabies virology, Rabies virus pathogenicity
- Abstract
Neuroinvasive viruses, such as alpha herpesviruses (αHV) and rabies virus (RABV), initially infect peripheral tissues, followed by invasion of the innervating axon termini. Virus particles must undergo long distance retrograde axonal transport to reach the neuron cell bodies in the peripheral or central nervous system (PNS/CNS). How virus particles hijack the axonal transport machinery and how PNS axons respond to and regulate infection are questions of significant interest. To track individual virus particles, we constructed a recombinant RABV expressing a P-mCherry fusion protein, derived from the virulent CVS-N2c strain. We studied retrograde RABV transport in the presence or absence of interferons (IFN) or protein synthesis inhibitors, both of which were reported previously to restrict axonal transport of αHV particles. Using neurons from rodent superior cervical ganglia grown in tri-chambers, we showed that axonal exposure to type I or type II IFN did not alter retrograde axonal transport of RABV. However, exposure of axons to emetine, a translation elongation inhibitor, blocked axonal RABV transport by a mechanism that was not dependent on protein synthesis inhibition. The minority of RABV particles that still moved retrograde in axons in the presence of emetine, moved with slower velocities and traveled shorter distances. Emetine's effect was specific to RABV, as transport of cellular vesicles was unchanged. These findings extend our understanding of how neuroinvasion is regulated in axons and point toward a role for emetine as an inhibitory modulator of RABV axonal transport., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2018
- Full Text
- View/download PDF
25. The Final (Oral Ebola) Vaccine Trial on Captive Chimpanzees?
- Author
-
Walsh PD, Kurup D, Hasselschwert DL, Wirblich C, Goetzmann JE, and Schnell MJ
- Subjects
- Administration, Oral, Animals, Antibodies, Neutralizing blood, Antibodies, Viral blood, Ebola Vaccines administration & dosage, Ebola Vaccines genetics, Ebolavirus genetics, Hemorrhagic Fever, Ebola prevention & control, Injections, Intramuscular, Pan troglodytes, Rabies virus genetics, Treatment Outcome, Vaccines, Synthetic administration & dosage, Vaccines, Synthetic genetics, Vaccines, Synthetic immunology, Drug Carriers, Ebola Vaccines immunology, Ebolavirus immunology, Hemorrhagic Fever, Ebola veterinary, Monkey Diseases prevention & control
- Abstract
Could new oral vaccine technologies protect endangered wildlife against a rising tide of infectious disease? We used captive chimpanzees to test oral delivery of a rabies virus (RABV) vectored vaccine against Ebola virus (EBOV), a major threat to wild chimpanzees and gorillas. EBOV GP and RABV GP-specific antibody titers increased exponentially during the trial, with rates of increase for six orally vaccinated chimpanzees very similar to four intramuscularly vaccinated controls. Chimpanzee sera also showed robust neutralizing activity against RABV and pseudo-typed EBOV. Vaccination did not induce serious health complications. Blood chemistry, hematologic, and body mass correlates of psychological stress suggested that, although sedation induced acute stress, experimental housing conditions did not induce traumatic levels of chronic stress. Acute behavioral and physiological responses to sedation were strongly correlated with immune responses to vaccination. These results suggest that oral vaccination holds great promise as a tool for the conservation of apes and other endangered tropical wildlife. They also imply that vaccine and drug trials on other captive species need to better account for the effects of stress on immune response.
- Published
- 2017
- Full Text
- View/download PDF
26. One-Health: a Safe, Efficient, Dual-Use Vaccine for Humans and Animals against Middle East Respiratory Syndrome Coronavirus and Rabies Virus.
- Author
-
Wirblich C, Coleman CM, Kurup D, Abraham TS, Bernbaum JG, Jahrling PB, Hensley LE, Johnson RF, Frieman MB, and Schnell MJ
- Subjects
- Animals, Coronavirus Infections prevention & control, Coronavirus Infections virology, Disease Models, Animal, Gene Expression Regulation, Viral, Humans, Immunization, Mice, Microbial Interactions, Middle East Respiratory Syndrome Coronavirus genetics, Rabies prevention & control, Rabies virology, Rabies virus genetics, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins immunology, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus immunology, Vaccines, Attenuated, Vaccines, Synthetic, Viral Proteins genetics, Viral Proteins immunology, Viral Vaccines administration & dosage, Viral Vaccines adverse effects, Viral Vaccines genetics, Virus Assembly, Coronavirus Infections immunology, Cross Protection immunology, Middle East Respiratory Syndrome Coronavirus immunology, Rabies immunology, Rabies virus immunology, Viral Vaccines immunology
- Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) emerged in 2012 and is a highly pathogenic respiratory virus. There are no treatment options against MERS-CoV for humans or animals, and there are no large-scale clinical trials for therapies against MERS-CoV. To address this need, we developed an inactivated rabies virus (RABV) that contains the MERS-CoV spike (S) protein expressed on its surface. Our initial recombinant vaccine, BNSP333-S, expresses a full-length wild-type MERS-CoV S protein; however, it showed significantly reduced viral titers compared to those of the parental RABV strain and only low-level incorporation of full-length MERS-CoV S into RABV particles. Therefore, we developed a RABV-MERS vector that contained the MERS-CoV S1 domain of the MERS-CoV S protein fused to the RABV G protein C terminus (BNSP333-S1). BNSP333-S1 grew to titers similar to those of the parental vaccine vector BNSP333, and the RABV G-MERS-CoV S1 fusion protein was efficiently expressed and incorporated into RABV particles. When we vaccinated mice, chemically inactivated BNSP333-S1 induced high-titer neutralizing antibodies. Next, we challenged both vaccinated mice and control mice with MERS-CoV after adenovirus transduction of the human dipeptidyl peptidase 4 (hDPP4) receptor and then analyzed the ability of mice to control MERS-CoV infection. Our results demonstrated that vaccinated mice were fully protected from the MERS-CoV challenge, as indicated by the significantly lower MERS-CoV titers and MERS-CoV and mRNA levels in challenged mice than those in unvaccinated controls. These data establish that an inactivated RABV-MERS S-based vaccine may be effective for use in animals and humans in areas where MERS-CoV is endemic., Importance: Rabies virus-based vectors have been proven to be efficient dual vaccines against rabies and emergent infectious diseases such as Ebola virus. Here we show that inactivated rabies virus particles containing the MERS-CoV S1 protein induce potent immune responses against MERS-CoV and RABV. This novel vaccine is easy to produce and may be useful to protect target animals, such as camels, as well as humans from deadly MERS-CoV and RABV infections. Our results indicate that this vaccine approach can prevent disease, and the RABV-based vaccine platform may be a valuable tool for timely vaccine development against emerging infectious diseases., (Copyright © 2017 American Society for Microbiology.)
- Published
- 2017
- Full Text
- View/download PDF
27. An Inactivated Rabies Virus-Based Ebola Vaccine, FILORAB1, Adjuvanted With Glucopyranosyl Lipid A in Stable Emulsion Confers Complete Protection in Nonhuman Primate Challenge Models.
- Author
-
Johnson RF, Kurup D, Hagen KR, Fisher C, Keshwara R, Papaneri A, Perry DL, Cooper K, Jahrling PB, Wang JT, Ter Meulen J, Wirblich C, and Schnell MJ
- Subjects
- Adjuvants, Immunologic, Animals, Antibodies, Viral immunology, Emulsions, Female, Hemorrhagic Fever, Ebola immunology, Hemorrhagic Fever, Ebola virology, Macaca fascicularis, Macaca mulatta, Male, Rabies immunology, Rabies virology, Rabies Vaccines immunology, Toll-Like Receptor 4 immunology, Vaccines, Inactivated immunology, Vaccines, Synthetic immunology, Ebola Vaccines immunology, Ebolavirus immunology, Glucosides immunology, Hemorrhagic Fever, Ebola prevention & control, Lipid A immunology, Rabies prevention & control, Rabies virus immunology
- Abstract
The 2013-2016 West African Ebola virus (EBOV) disease outbreak was the largest filovirus outbreak to date. Over 28 000 suspected, probable, or confirmed cases have been reported, with a 53% case-fatality rate. The magnitude and international impact of this EBOV outbreak has highlighted the urgent need for a safe and efficient EBOV vaccine. To this end, we demonstrate the immunogenicity and protective efficacy of FILORAB1, a recombinant, bivalent, inactivated rabies virus-based EBOV vaccine, in rhesus and cynomolgus monkeys. Our results demonstrate that the use of the synthetic Toll-like receptor 4 agonist glucopyranosyl lipid A in stable emulsion (GLA-SE) as an adjuvant increased the efficacy of FILORAB1 to 100% protection against lethal EBOV challenge, with no to mild clinical signs of disease. Furthermore, all vaccinated subjects developed protective anti-rabies virus antibody titers. Taken together, these results support further development of FILORAB1/GLA-SE as an effective preexposure EBOV vaccine., (Published by Oxford University Press for the Infectious Diseases Society of America 2016. This work is written by (a) US Government employee(s) and is in the public domain in the US.)
- Published
- 2016
- Full Text
- View/download PDF
28. Rabies Virus CVS-N2c(ΔG) Strain Enhances Retrograde Synaptic Transfer and Neuronal Viability.
- Author
-
Reardon TR, Murray AJ, Turi GF, Wirblich C, Croce KR, Schnell MJ, Jessell TM, and Losonczy A
- Subjects
- Action Potentials genetics, Animals, Cells, Cultured, Electric Stimulation, Glycoproteins genetics, Humans, Luminescent Proteins genetics, Luminescent Proteins metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Neural Pathways physiology, Neuroblastoma pathology, Neurons virology, Optogenetics, Protein Transport, Viral Envelope Proteins, Glycoproteins deficiency, Nerve Net physiology, Neurons physiology, Rabies virus physiology
- Abstract
Virally based transsynaptic tracing technologies are powerful experimental tools for neuronal circuit mapping. The glycoprotein-deletion variant of the SAD-B19 vaccine strain rabies virus (RABV) has been the reagent of choice in monosynaptic tracing, since it permits the mapping of synaptic inputs to genetically marked neurons. Since its introduction, new helper viruses and reagents that facilitate complementation have enhanced the efficiency of SAD-B19(ΔG) transsynaptic transfer, but there has been little focus on improvements to the core RABV strain. Here we generate a new deletion mutant strain, CVS-N2c(ΔG), and examine its neuronal toxicity and efficiency in directing retrograde transsynaptic transfer. We find that by comparison with SAD-B19(ΔG), the CVS-N2c(ΔG) strain exhibits a reduction in neuronal toxicity and a marked enhancement in transsynaptic neuronal transfer. We conclude that the CVS-N2c(ΔG) strain provides a more effective means of mapping neuronal circuitry and of monitoring and manipulating neuronal activity in vivo in the mammalian CNS., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
29. Preclinical Development of Inactivated Rabies Virus-Based Polyvalent Vaccine Against Rabies and Filoviruses.
- Author
-
Willet M, Kurup D, Papaneri A, Wirblich C, Hooper JW, Kwilas SA, Keshwara R, Hudacek A, Beilfuss S, Rudolph G, Pommerening E, Vos A, Neubert A, Jahrling P, Blaney JE, Johnson RF, and Schnell MJ
- Subjects
- Animals, Antibodies, Viral immunology, Antibody Formation immunology, Chlorocebus aethiops, Drug Evaluation, Preclinical methods, Ebola Vaccines immunology, Ebolavirus immunology, Glycoproteins immunology, Hemorrhagic Fever, Ebola immunology, Hemorrhagic Fever, Ebola virology, Macaca fascicularis, Marburgvirus immunology, Mice, Mice, Inbred C57BL, Rabies virology, Sudan, Vaccination methods, Vero Cells, Filoviridae immunology, Rabies immunology, Rabies Vaccines immunology, Rabies virus immunology, Vaccines, Inactivated immunology
- Abstract
We previously described the generation of a novel Ebola virus (EBOV) vaccine based on inactivated rabies virus (RABV) containing EBOV glycoprotein (GP) incorporated in the RABV virion. Our results demonstrated safety, immunogenicity, and protective efficacy in mice and nonhuman primates (NHPs). Protection against viral challenge depended largely on the quality of the humoral immune response against EBOV GP.Here we present the extension and improvement of this vaccine by increasing the amount of GP incorporation into virions via GP codon-optimization as well as the addition of Sudan virus (SUDV) and Marburg virus (MARV) GP containing virions. Immunogenicity studies in mice indicate similar immune responses for both SUDV GP and MARV GP compared to EBOV GP. Immunizing mice with multiple antigens resulted in immune responses similar to immunization with a single antigen. Moreover, immunization of NHP with the new inactivated RABV EBOV vaccine resulted in high titer neutralizing antibody levels and 100% protection against lethal EBOV challenge when applied with adjuvant.Our results indicate that an inactivated polyvalent vaccine against RABV filoviruses is achievable. Finally, the novel vaccines are produced on approved VERO cells and a clinical grade RABV/EBOV vaccine for human trials has been produced., (Published by Oxford University Press on behalf of the Infectious Diseases Society of America 2015. This work is written by (a) US Government employee(s) and is in the public domain in the US.)
- Published
- 2015
- Full Text
- View/download PDF
30. Focal adhesion kinase is involved in rabies virus infection through its interaction with viral phosphoprotein P.
- Author
-
Fouquet B, Nikolic J, Larrous F, Bourhy H, Wirblich C, Lagaudrière-Gesbert C, and Blondel D
- Subjects
- Animals, Cell Line, DNA Mutational Analysis, Humans, Immunoprecipitation, Inclusion Bodies, Viral chemistry, Inclusion Bodies, Viral virology, Microscopy, Confocal, Molecular Chaperones, Mutagenesis, Site-Directed, Protein Binding, Two-Hybrid System Techniques, Focal Adhesion Protein-Tyrosine Kinases metabolism, Host-Pathogen Interactions, Phosphoproteins metabolism, Protein Interaction Mapping, Rabies virus physiology, Viral Structural Proteins metabolism, Virus Replication
- Abstract
Unlabelled: The rabies virus (RABV) phosphoprotein P is a multifunctional protein: it plays an essential role in viral transcription and replication, and in addition, RABV P has been identified as an interferon antagonist. Here, a yeast two-hybrid screen revealed that RABV P interacts with the focal adhesion kinase (FAK). The binding involved the 106-to-131 domain, corresponding to the dimerization domain of P and the C-terminal domain of FAK containing the proline-rich domains PRR2 and PRR3. The P-FAK interaction was confirmed in infected cells by coimmunoprecipitation and colocalization of FAK with P in Negri bodies. By alanine scanning, we identified a single mutation in the P protein that abolishes this interaction. The mutant virus containing a substitution of Ala for Arg in position 109 in P (P.R109A), which did not interact with FAK, is affected at a posttranscriptional step involving protein synthesis and viral RNA replication. Furthermore, FAK depletion inhibited viral protein expression in infected cells. This provides the first evidence of an interaction of RABV with FAK that positively regulates infection., Importance: Rabies virus exhibits a small genome that encodes a limited number of viral proteins. To maintain efficient virus replication, some of them are multifunctional, such as the phosphoprotein P. We and others have shown that P establishes complex networks of interactions with host cell components. These interactions have revealed much about the role of P and about host-pathogen interactions in infected cells. Here, we identified another cellular partner of P, the focal adhesion kinase (FAK). Our data shed light on the implication of FAK in RABV infection and provide evidence that P-FAK interaction has a proviral function., (Copyright © 2015, American Society for Microbiology. All Rights Reserved.)
- Published
- 2015
- Full Text
- View/download PDF
31. Rhabdovirus-based vaccine platforms against henipaviruses.
- Author
-
Kurup D, Wirblich C, Feldmann H, Marzi A, and Schnell MJ
- Subjects
- Administration, Intranasal, Animals, Antibodies, Neutralizing blood, Antibodies, Viral blood, Drug Carriers administration & dosage, Drug-Related Side Effects and Adverse Reactions, Hendra Virus genetics, Henipavirus Infections immunology, Injections, Intramuscular, Mice, Mice, Inbred BALB C, Vaccination methods, Vaccines, Attenuated administration & dosage, Vaccines, Attenuated genetics, Vaccines, Attenuated immunology, Vaccines, Inactivated administration & dosage, Vaccines, Inactivated genetics, Vaccines, Inactivated immunology, Vaccines, Synthetic administration & dosage, Vaccines, Synthetic genetics, Vaccines, Synthetic immunology, Viral Envelope Proteins genetics, Viral Vaccines administration & dosage, Viral Vaccines genetics, Genetic Vectors, Hendra Virus immunology, Henipavirus Infections prevention & control, Rabies virus genetics, Vesiculovirus genetics, Viral Envelope Proteins immunology, Viral Vaccines immunology
- Abstract
Unlabelled: The emerging zoonotic pathogens Hendra virus (HeV) and Nipah virus (NiV) are in the genus Henipavirus in the family Paramyxoviridae. HeV and NiV infections can be highly fatal to humans and livestock. The goal of this study was to develop candidate vaccines against henipaviruses utilizing two well-established rhabdoviral vaccine vector platforms, recombinant rabies virus (RABV) and recombinant vesicular stomatitis virus (VSV), expressing either the codon-optimized or the wild-type (wt) HeV glycoprotein (G) gene. The RABV vector expressing the codon-optimized HeV G showed a 2- to 3-fold increase in incorporation compared to the RABV vector expressing wt HeV G. There was no significant difference in HeV G incorporation in the VSV vectors expressing either wt or codon-optimized HeV G. Mice inoculated intranasally with any of these live recombinant viruses showed no signs of disease, including weight loss, indicating that HeV G expression and incorporation did not increase the neurotropism of the vaccine vectors. To test the immunogenicity of the vaccine candidates, we immunized mice intramuscularly with either one dose of the live vaccines or 3 doses of 10 μg chemically inactivated viral particles. Increased codon-optimized HeV G incorporation into RABV virions resulted in higher antibody titers against HeV G compared to inactivated RABV virions expressing wt HeV G. The live VSV vectors induced more HeV G-specific antibodies as well as higher levels of HeV neutralizing antibodies than the RABV vectors. In the case of killed particles, HeV neutralizing serum titers were very similar between the two platforms. These results indicated that killed RABV with codon-optimized HeV G should be the vector of choice as a dual vaccine in areas where rabies is endemic., Importance: Scientists have been tracking two new viruses carried by the Pteropid fruit bats: Hendra virus (HeV) and Nipah virus (NiV). Both viruses can be fatal to humans and also pose a serious risk to domestic animals. A recent escalation in the frequency of outbreaks has increased the need for a vaccine that prevents HeV and NiV infections. In this study, we performed an extensive comparison of live and killed particles of two recombinant rhabdoviral vectors, rabies virus and vesicular stomatitis virus (VSV), expressing wild-type or codon-optimized HeV glycoprotein, with the goal of developing a candidate vaccine against HeV. Based on our data from the presented mouse immunogenicity studies, we conclude that a killed RABV vaccine would be highly effective against HeV infections and would make an excellent vaccine candidate in areas where both RABV and henipaviruses pose a threat to human health., (Copyright © 2015, American Society for Microbiology. All Rights Reserved.)
- Published
- 2015
- Full Text
- View/download PDF
32. Alanine scanning of the rabies virus glycoprotein antigenic site III using recombinant rabies virus: implication for post-exposure treatment.
- Author
-
Papaneri AB, Wirblich C, Marissen WE, and Schnell MJ
- Subjects
- Alanine genetics, Animals, Antibodies, Monoclonal therapeutic use, Antibodies, Neutralizing immunology, Antibodies, Neutralizing therapeutic use, Antibodies, Viral immunology, Antigens, Viral immunology, Binding Sites, Antibody, Cell Line, Cricetinae, Glycoproteins immunology, Humans, Mutagenesis, Site-Directed, Neutralization Tests, Rabies Vaccines therapeutic use, Viral Envelope Proteins genetics, Viral Envelope Proteins immunology, Antibodies, Monoclonal immunology, Antigens, Viral genetics, Glycoproteins genetics, Post-Exposure Prophylaxis, Rabies therapy
- Abstract
The safety and availability of the human polyclonal sera that is currently utilized for post-exposure treatment (PET) of rabies virus (RABV) infection remain a concern. Recombinant monoclonal antibodies have been postulated as suitable alternatives by WHO. To this extent, CL184, the RABV human antibody combination comprising monoclonal antibodies (mAbs) CR57 and CR4098, has been developed and has delivered promising clinical data to support its use for RABV PET. For this fully human IgG1 cocktail, mAbs CR57 and CR4098 are produced in the PER.C6 human cell line and combined in equal amounts in the final product. During preclinical evaluation, CR57 was shown to bind to antigenic site I whereas CR4098 neutralization was influenced by a mutation of position 336 (N336) located within antigenic site III. Here, alanine scanning was used to analyze the influence of mutations within the potential binding site for CR4098, antigenic site III, in order to evaluate the possibility of mutated rabies viruses escaping neutralization. For this approach, twenty flanking amino acids (10 upstream and 10 downstream) of the RABV glycoprotein (G) asparagine (N336) were exchanged to alanine (or serine, if already alanine) by site-directed mutagenesis. Analysis of G expression revealed four of the twenty mutant Gs to be non-functional, as shown by their lack of cell surface expression, which is a requirement for the production of infectious RABV. Therefore, these mutants were excluded from further study. The remaining sixteen mutants were introduced in an infectious clone of RABV, and recombinant RABVs (rRABVs) were recovered and utilized for in vitro neutralization assays. All of the viruses were effectively neutralized by CR4098 as well as by CR57, indicating that single amino acid exchanges in this region does not affect the broad neutralizing capability of the CL184 mAb combination., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
33. Comparison of Heterologous Prime-Boost Strategies against Human Immunodeficiency Virus Type 1 Gag Using Negative Stranded RNA Viruses.
- Author
-
Lawrence TM, Wanjalla CN, Gomme EA, Wirblich C, Gatt A, Carnero E, García-Sastre A, Lyles DS, McGettigan JP, and Schnell MJ
- Subjects
- Animals, CD8-Positive T-Lymphocytes immunology, DNA, Recombinant genetics, Female, Genetic Vectors genetics, Mice, Mice, Inbred BALB C, HIV-1 immunology, Immunization, Secondary methods, RNA Viruses genetics, gag Gene Products, Human Immunodeficiency Virus genetics, gag Gene Products, Human Immunodeficiency Virus immunology
- Abstract
This study analyzed a heterologous prime-boost vaccine approach against HIV-1 using three different antigenically unrelated negative-stranded viruses (NSV) expressing HIV-1 Gag as vaccine vectors: rabies virus (RABV), vesicular stomatitis virus (VSV) and Newcastle disease virus (NDV). We hypothesized that this approach would result in more robust cellular immune responses than those achieved with the use of any of the vaccines alone in a homologous prime-boost regimen. To this end, we primed BALB/c mice with each of the NSV-based vectors. Primed mice were rested for thirty-five days after which we administered a second immunization with the same or heterologous NSV-Gag viruses. The magnitude and quality of the Gag-specific CD8(+) T cells in response to these vectors post boost were measured. In addition, we performed challenge experiments using vaccinia virus expressing HIV-1 Gag (VV-Gag) thirty-three days after the boost inoculation. Our results showed that the choice of the vaccine used for priming was important for the detected Gag-specific CD8(+) T cell recall responses post boost and that NDV-Gag appeared to result in a more robust recall of CD8(+) T cell responses independent of the prime vaccine used. However, the different prime-boost strategies were not distinct for the parameters studied in the challenge experiments using VV-Gag but did indicate some benefits compared to single immunizations. Taken together, our data show that NSV vectors can individually stimulate HIV-Gag specific CD8(+) T cells that are effectively recalled by other NSV vectors in a heterologous prime-boost approach. These results provide evidence that RABV, VSV and NDV can be used in combination to develop vaccines needing prime-boost regimens to stimulate effective immune responses.
- Published
- 2013
- Full Text
- View/download PDF
34. Antibody quality and protection from lethal Ebola virus challenge in nonhuman primates immunized with rabies virus based bivalent vaccine.
- Author
-
Blaney JE, Marzi A, Willet M, Papaneri AB, Wirblich C, Feldmann F, Holbrook M, Jahrling P, Feldmann H, and Schnell MJ
- Subjects
- Animals, Female, Macaca mulatta, Male, Mice, Antibodies, Viral immunology, Ebola Vaccines genetics, Ebola Vaccines immunology, Ebola Vaccines pharmacology, Ebolavirus genetics, Ebolavirus immunology, Hemorrhagic Fever, Ebola immunology, Hemorrhagic Fever, Ebola prevention & control, Immunoglobulin G immunology, Rabies Vaccines genetics, Rabies Vaccines immunology, Rabies Vaccines pharmacology, Rabies virus genetics, Rabies virus immunology, Viral Matrix Proteins genetics, Viral Matrix Proteins immunology, Viral Matrix Proteins pharmacology
- Abstract
We have previously described the generation of a novel Ebola virus (EBOV) vaccine platform based on (a) replication-competent rabies virus (RABV), (b) replication-deficient RABV, or (c) chemically inactivated RABV expressing EBOV glycoprotein (GP). Mouse studies demonstrated safety, immunogenicity, and protective efficacy of these live or inactivated RABV/EBOV vaccines. Here, we evaluated these vaccines in nonhuman primates. Our results indicate that all three vaccines do induce potent immune responses against both RABV and EBOV, while the protection of immunized animals against EBOV was largely dependent on the quality of humoral immune response against EBOV GP. We also determined if the induced antibodies against EBOV GP differ in their target, affinity, or the isotype. Our results show that IgG1-biased humoral responses as well as high levels of GP-specific antibodies were beneficial for the control of EBOV infection after immunization. These results further support the concept that a successful EBOV vaccine needs to induce strong antibodies against EBOV. We also showed that a dual vaccine against RABV and filoviruses is achievable; therefore addressing concerns for the marketability of this urgently needed vaccine.
- Published
- 2013
- Full Text
- View/download PDF
35. A replication-deficient rabies virus vaccine expressing Ebola virus glycoprotein is highly attenuated for neurovirulence.
- Author
-
Papaneri AB, Wirblich C, Cann JA, Cooper K, Jahrling PB, Schnell MJ, and Blaney JE
- Subjects
- Animals, Animals, Newborn, Brain pathology, Brain virology, Disease Models, Animal, Ebola Vaccines genetics, Female, Gene Deletion, Genes, Viral, Immunohistochemistry, Mice, Mice, Inbred BALB C, Mice, SCID, Rabies Vaccines genetics, Real-Time Polymerase Chain Reaction, Vaccines, Attenuated adverse effects, Vaccines, Attenuated genetics, Vaccines, Attenuated immunology, Viral Load, Virulence, Ebola Vaccines adverse effects, Ebola Vaccines immunology, Rabies Vaccines adverse effects, Rabies Vaccines immunology
- Abstract
We are developing inactivated and live-attenuated rabies virus (RABV) vaccines expressing Ebola virus (EBOV) glycoprotein for use in humans and endangered wildlife, respectively. Here, we further characterize the pathogenesis of the live-attenuated RABV/EBOV vaccine candidates in mice in an effort to define their growth properties and potential for safety. RABV vaccines expressing GP (RV-GP) or a replication-deficient derivative with a deletion of the RABV G gene (RVΔG-GP) are both avirulent after intracerebral inoculation of adult mice. Furthermore, RVΔG-GP is completely avirulent upon intracerebral inoculation of suckling mice unlike parental RABV vaccine or RV-GP. Analysis of RVΔG-GP in the brain by quantitative PCR, determination of virus titer, and immunohistochemistry indicated greatly restricted virus replication. In summary, our findings indicate that RV-GP retains the attenuation phenotype of the live-attenuated RABV vaccine, and RVΔG-GP would appear to be an even safer alternative for use in wildlife or consideration for human use., (Published by Elsevier Inc.)
- Published
- 2012
- Full Text
- View/download PDF
36. Further characterization of the immune response in mice to inactivated and live rabies vaccines expressing Ebola virus glycoprotein.
- Author
-
Papaneri AB, Wirblich C, Cooper K, Jahrling PB, Schnell MJ, and Blaney JE
- Subjects
- Animals, Antibodies, Viral blood, Ebolavirus immunology, Immunity, Cellular, Immunity, Humoral, Interferon-gamma immunology, Mice, Mice, Inbred BALB C, T-Lymphocytes immunology, Vaccines, Inactivated immunology, Antibody Specificity, Ebola Vaccines immunology, Hemorrhagic Fever, Ebola prevention & control, Membrane Glycoproteins immunology, Rabies Vaccines immunology, Viral Matrix Proteins immunology
- Abstract
We have previously developed (a) replication-competent, (b) replication-deficient, and (c) chemically inactivated rabies virus (RABV) vaccines expressing Ebola virus (EBOV) glycoprotein (GP) that induce humoral immunity against each virus and confer protection from both lethal RABV and mouse-adapted EBOV challenge in mice. Here, we expand our investigation of the immunogenic properties of these bivalent vaccines in mice. Both live and killed vaccines induced primary EBOV GP-specific T-cells and a robust recall response as measured by interferon-γ ELISPOT assay. In addition to cellular immunity, an effective filovirus vaccine will likely require a multivalent humoral immune response against multiple virus species. As a proof-of-principle experiment, we demonstrated that inactivated RV-GP could be formulated with another inactivated RABV vaccine expressing the nontoxic fragment of botulinum neurotoxin A heavy chain (HC50) without a reduction in immunity to each component. Finally, we demonstrated that humoral immunity to GP could be induced by immunization of mice with inactivated RV-GP in the presence of pre-existing immunity to RABV. The ability of these novel vaccines to induce strong humoral and cellular immunity indicates that they should be further evaluated in additional animal models of infection., (Published by Elsevier Ltd.)
- Published
- 2012
- Full Text
- View/download PDF
37. Targeted single-neuron infection with rabies virus for transneuronal multisynaptic tracing.
- Author
-
Nguyen TD, Wirblich C, Aizenman E, Schnell MJ, Strick PL, and Kandler K
- Subjects
- Animals, Cells, Cultured, Cerebral Cortex cytology, Embryo, Mammalian, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Organ Culture Techniques, Rabies virus genetics, Rats, Rats, Sprague-Dawley, Transfection, Neurons metabolism, Neurons virology, Rabies virus physiology
- Abstract
The transynaptic and retrograde spread of rabies virus make it an efficient and robust transneuronal tracer, capable of revealing connectivity patterns of multisynaptic, neuronal circuits with great detail. Current techniques begin by infecting many neurons simultaneously, from which higher-order neurons are then labeled sequentially in time. Here we report on a method that can initially infect a single neuron-of-choice, allowing for greater precision and specificity of labeled circuits., (Copyright © 2012 Elsevier B.V. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
38. A role for granulocyte-macrophage colony-stimulating factor in the regulation of CD8(+) T cell responses to rabies virus.
- Author
-
Wanjalla CN, Goldstein EF, Wirblich C, and Schnell MJ
- Subjects
- Adjuvants, Immunologic genetics, Animals, Antigen-Presenting Cells immunology, Female, Granulocyte-Macrophage Colony-Stimulating Factor genetics, HIV Infections immunology, HIV Infections virology, Humans, Mice, Mice, Inbred BALB C, Rabies virology, Rabies virus genetics, Spleen immunology, gag Gene Products, Human Immunodeficiency Virus genetics, gag Gene Products, Human Immunodeficiency Virus immunology, CD8-Positive T-Lymphocytes immunology, Granulocyte-Macrophage Colony-Stimulating Factor immunology, Rabies immunology, Rabies virus immunology
- Abstract
Inflammatory cytokines have a significant role in altering the innate and adaptive arms of immune responses. Here, we analyzed the effect of GM-CSF on a RABV-vaccine vector co-expressing HIV-1 Gag. To this end, we immunized mice with RABV expressing HIV-1 Gag and GM-CSF and analyzed the primary and recall CD8(+) T cell responses. We observed a statistically significant increase in antigen presenting cells (APCs) in the spleen and draining lymph nodes in response to GM-CSF. Despite the increase in APCs, the primary and memory anti HIV-1 CD8(+) T cell response was significantly lower. This was partly likely due to lower levels of proliferation in the spleen. Animals treated with GM-CSF neutralizing antibodies restored the CD8(+) T cell response. These data define a role of GM-CSF expression, in the regulation of the CD8(+) T cell immune responses against RABV and has implications in the use of GM-CSF as a molecular adjuvant in vaccine development., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
39. Immune clearance of attenuated rabies virus results in neuronal survival with altered gene expression.
- Author
-
Gomme EA, Wirblich C, Addya S, Rall GF, and Schnell MJ
- Subjects
- Animals, Brain immunology, Brain pathology, Cell Survival, Gene Expression, Green Fluorescent Proteins genetics, Integrases genetics, Mice, Neurons immunology, Neurons virology, Rabies genetics, Rabies pathology, Rabies virology, Rabies virus pathogenicity, Real-Time Polymerase Chain Reaction, Transcriptome, Brain virology, Neurons physiology, Rabies immunology, Rabies virus immunology
- Abstract
Rabies virus (RABV) is a highly neurotropic pathogen that typically leads to mortality of infected animals and humans. The precise etiology of rabies neuropathogenesis is unknown, though it is hypothesized to be due either to neuronal death or dysfunction. Analysis of human brains post-mortem reveals surprisingly little tissue damage and neuropathology considering the dramatic clinical symptomology, supporting the neuronal dysfunction model. However, whether or not neurons survive infection and clearance and, provided they do, whether they are functionally restored to their pre-infection phenotype has not been determined in vivo for RABV, or any neurotropic virus. This is due, in part, to the absence of a permanent "mark" on once-infected cells that allow their identification long after viral clearance. Our approach to study the survival and integrity of RABV-infected neurons was to infect Cre reporter mice with recombinant RABV expressing Cre-recombinase (RABV-Cre) to switch neurons constitutively expressing tdTomato (red) to expression of a Cre-inducible EGFP (green), permanently marking neurons that had been infected in vivo. We used fluorescence microscopy and quantitative real-time PCR to measure the survival of neurons after viral clearance; we found that the vast majority of RABV-infected neurons survive both infection and immunological clearance. We were able to isolate these previously infected neurons by flow cytometry and assay their gene expression profiles compared to uninfected cells. We observed transcriptional changes in these "cured" neurons, predictive of decreased neurite growth and dysregulated microtubule dynamics. This suggests that viral clearance, though allowing for survival of neurons, may not restore them to their pre-infection functionality. Our data provide a proof-of-principle foundation to re-evaluate the etiology of human central nervous system diseases of unknown etiology: viruses may trigger permanent neuronal damage that can persist or progress in the absence of sustained viral antigen.
- Published
- 2012
- Full Text
- View/download PDF
40. Inactivated or live-attenuated bivalent vaccines that confer protection against rabies and Ebola viruses.
- Author
-
Blaney JE, Wirblich C, Papaneri AB, Johnson RF, Myers CJ, Juelich TL, Holbrook MR, Freiberg AN, Bernbaum JG, Jahrling PB, Paragas J, and Schnell MJ
- Subjects
- Animals, Antibodies, Viral blood, Brain virology, Disease Models, Animal, Ebola Vaccines administration & dosage, Ebola Vaccines adverse effects, Ebola Vaccines genetics, Ebolavirus genetics, Ebolavirus immunology, Hemorrhagic Fever, Ebola prevention & control, Mice, Mice, Inbred BALB C, Rabies prevention & control, Rabies Vaccines administration & dosage, Rabies Vaccines adverse effects, Rabies Vaccines genetics, Rabies virus genetics, Rabies virus immunology, Rodent Diseases prevention & control, Vaccines, Attenuated administration & dosage, Vaccines, Attenuated adverse effects, Vaccines, Attenuated genetics, Vaccines, Attenuated immunology, Vaccines, Inactivated administration & dosage, Vaccines, Inactivated adverse effects, Vaccines, Inactivated genetics, Vaccines, Inactivated immunology, Vaccines, Synthetic administration & dosage, Vaccines, Synthetic adverse effects, Vaccines, Synthetic genetics, Vaccines, Synthetic immunology, Virulence, Ebola Vaccines immunology, Rabies Vaccines immunology
- Abstract
The search for a safe and efficacious vaccine for Ebola virus continues, as no current vaccine candidate is nearing licensure. We have developed (i) replication-competent, (ii) replication-deficient, and (iii) chemically inactivated rabies virus (RABV) vaccines expressing Zaire Ebola virus (ZEBOV) glycoprotein (GP) by a reverse genetics system based on the SAD B19 RABV wildlife vaccine. ZEBOV GP is efficiently expressed by these vaccine candidates and is incorporated into virions. The vaccine candidates were avirulent after inoculation of adult mice, and viruses with a deletion in the RABV glycoprotein had greatly reduced neurovirulence after intracerebral inoculation in suckling mice. Immunization with live or inactivated RABV vaccines expressing ZEBOV GP induced humoral immunity against each virus and conferred protection from both lethal RABV and EBOV challenge in mice. The bivalent RABV/ZEBOV vaccines described here have several distinct advantages that may speed the development of inactivated vaccines for use in humans and potentially live or inactivated vaccines for use in nonhuman primates at risk of EBOV infection in endemic areas.
- Published
- 2011
- Full Text
- View/download PDF
41. Rabies virus as a research tool and viral vaccine vector.
- Author
-
Gomme EA, Wanjalla CN, Wirblich C, and Schnell MJ
- Subjects
- Animals, Genetic Engineering methods, Genetics, Microbial methods, Humans, Drug Carriers, Genetic Vectors, Rabies virus genetics, Rabies virus immunology, Viral Vaccines genetics, Viral Vaccines immunology
- Abstract
Until recently, single-stranded negative sense RNA viruses (ssNSVs) were one of only a few important human viral pathogens, which could not be created from cDNA. The inability to manipulate their genomes hindered their detailed genetic analysis. A key paper from Conzelmann's laboratory in 1994 changed this with the publication of a method to recover rabies virus (RABV) from cDNA. This discovery not only dramatically changed the broader field of ssNSV biology but also opened a whole new avenue for studying RABV pathogenicity, developing novel RABV vaccines as well a new generation of RABV-based vaccine vectors, and creating research tools important in neuroscience such as neuronal tracing., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
42. Rabies virus (RV) glycoprotein expression levels are not critical for pathogenicity of RV.
- Author
-
Wirblich C and Schnell MJ
- Subjects
- Animals, Antigens, Viral genetics, Cell Line, Codon, Disease Models, Animal, Glycoproteins genetics, Male, Mice, Molecular Sequence Data, Neurons virology, Rabies pathology, Rabies virology, Rabies virus genetics, Sequence Analysis, DNA, Survival Analysis, Viral Envelope Proteins genetics, Antigens, Viral biosynthesis, Gene Expression, Glycoproteins biosynthesis, Rabies virus pathogenicity, Viral Envelope Proteins biosynthesis
- Abstract
Previous comparisons of different rabies virus (RV) strains suggested an inverse relationship between pathogenicity and the amount of glycoprotein produced in infected cells. In order to provide more insight into this relationship, we pursued an experimental approach that allowed us to alter the glycoprotein expression level without altering the glycoprotein sequence, thereby eliminating the contribution of amino acid changes to differences in viral virulence. To this end, we constructed an infectious clone of the highly pathogenic rabies virus strain CVS-N2c and replaced its cognate glycoprotein gene with synthetic versions in which silent mutations were introduced to replace wild-type codons with the most or least frequently used synonymous codons. A recombinant N2c variant containing the fully codon-optimized G gene and three variants carrying a partially codon-deoptimized G gene were recovered on mouse neuroblastoma cells and shown to express 2- to 3-fold more and less glycoprotein, respectively, than wild-type N2c. Pathogenicity studies in mice revealed the WT-N2c virus to be the most pathogenic strain. Variants containing partially codon-deoptimized glycoprotein genes or the codon-optimized gene were less pathogenic than WT-N2c but still caused significant mortality. We conclude that the expression level of the glycoprotein gene does have an impact on pathogenicity but is not a dominant factor that determines pathogenicity. Thus, strategies such as changes in codon usage that aim solely at altering the expression level of the glycoprotein gene do not suffice to render a pathogenic rabies virus apathogenic and are not a viable and safe approach for attenuation of a pathogenic strain.
- Published
- 2011
- Full Text
- View/download PDF
43. Rabies virus infection induces type I interferon production in an IPS-1 dependent manner while dendritic cell activation relies on IFNAR signaling.
- Author
-
Faul EJ, Wanjalla CN, Suthar MS, Gale M, Wirblich C, and Schnell MJ
- Subjects
- Animals, Dendritic Cells virology, Mice, Mice, Inbred Strains, Receptor, Interferon alpha-beta metabolism, Signal Transduction immunology, Adaptor Proteins, Signal Transducing immunology, Dendritic Cells immunology, Interferon Type I biosynthesis, Rabies immunology, Receptor, Interferon alpha-beta immunology
- Abstract
As with many viruses, rabies virus (RABV) infection induces type I interferon (IFN) production within the infected host cells. However, RABV has evolved mechanisms by which to inhibit IFN production in order to sustain infection. Here we show that RABV infection of dendritic cells (DC) induces potent type I IFN production and DC activation. Although DCs are infected by RABV, the viral replication is highly suppressed in DCs, rendering the infection non-productive. We exploited this finding in bone marrow derived DCs (BMDC) in order to differentiate which pattern recognition receptor(s) (PRR) is responsible for inducing type I IFN following infection with RABV. Our results indicate that BMDC activation and type I IFN production following a RABV infection is independent of TLR signaling. However, IPS-1 is essential for both BMDC activation and IFN production. Interestingly, we see that the BMDC activation is primarily due to signaling through the IFNAR and only marginally induced by the initial infection. To further identify the receptor recognizing RABV infection, we next analyzed BMDC from Mda-5-/- and RIG-I-/- mice. In the absence of either receptor, there is a significant decrease in BMDC activation at 12h post infection. However, only RIG-I-/- cells exhibit a delay in type I IFN production. In order to determine the role that IPS-1 plays in vivo, we infected mice with pathogenic RABV. We see that IPS-1-/- mice are more susceptible to infection than IPS-1+/+ mice and have a significantly increased incident of limb paralysis.
- Published
- 2010
- Full Text
- View/download PDF
44. The cell biology of rabies virus: using stealth to reach the brain.
- Author
-
Schnell MJ, McGettigan JP, Wirblich C, and Papaneri A
- Subjects
- Animals, Humans, Models, Biological, Rabies virus pathogenicity, Viral Proteins physiology, Virulence Factors physiology, Brain virology, Peripheral Nervous System virology, Rabies virus physiology, Spinal Cord virology
- Abstract
Rabies virus, the prototypical neurotropic virus, causes one of the most lethal zoonotic diseases. According to official estimates, over 55,000 people die of the disease annually, but this is probably a severe underestimation. A combination of virulence factors enables the virus to enter neurons at peripheral sites and travel through the spinal cord to the brain of the infected host, where it often induces aggression that facilitates the transfer of the virus to a new host. This Review summarizes the current knowledge of the replication cycle of rabies virus and virus- host cell interactions, both of which are fundamental elements in our quest to understand the life cycle of rabies virus and the pathogenesis of rabies.
- Published
- 2010
- Full Text
- View/download PDF
45. PPEY motif within the rabies virus (RV) matrix protein is essential for efficient virion release and RV pathogenicity.
- Author
-
Wirblich C, Tan GS, Papaneri A, Godlewski PJ, Orenstein JM, Harty RN, and Schnell MJ
- Subjects
- Amino Acid Motifs, Amino Acid Sequence, Animals, DNA Primers chemistry, Mice, Mice, Inbred C57BL, Microscopy, Electron, Molecular Sequence Data, Mutation, Protein Structure, Tertiary, RNA chemistry, Rabies virus genetics, Sequence Homology, Amino Acid, Viral Matrix Proteins chemistry, Rabies virus metabolism, Virion chemistry
- Abstract
Late (L) domains containing the highly conserved sequence PPXY were first described for retroviruses, and later research confirmed their conservation and importance for efficient budding of several negative-stranded RNA viruses. Rabies virus (RV), a member of the Rhabdoviridae family, contains the sequence PPEY (amino acids 35 to 38) within the N terminus of the matrix (M) protein, but the functions of this potential L-domain in the viral life cycle, viral pathogenicity, and immunogenicity have not been established. Here we constructed a series of recombinant RVs containing mutations within the PPEY motif and analyzed their effects on viral replication and RV pathogenicity. Our results indicate that the first proline at position 35 is the most important for viral replication, whereas P36 and Y38 have a lesser but still noticeable impact. The reduction in viral replication was most likely due to inhibition of virion release, because initially no major impact on RV RNA synthesis was observed. In addition, results from electron microscopy demonstrated that the M4A mutant virus (PPEY-->SAEA) displayed a more cell-associated phenotype than that of wild-type RV. Furthermore, all mutations within the PPEY motif resulted in reduced spread of the recombinant RVs as indicated by a reduction in focus size. Importantly, recombinant PPEY L-domain mutants were highly attenuated in mice yet still elicited potent antibody responses against RV G protein that were as high as those observed after infection with wild-type virus. Our data indicate that the RV PPEY motif has L-domain activity essential for efficient virus production and pathogenicity but is not essential for immunogenicity and thus can be targeted to increase the safety of rabies vaccine vectors.
- Published
- 2008
- Full Text
- View/download PDF
46. Nonstructural protein 3 of bluetongue virus assists virus release by recruiting ESCRT-I protein Tsg101.
- Author
-
Wirblich C, Bhattacharya B, and Roy P
- Subjects
- DNA-Binding Proteins chemistry, Endosomal Sorting Complexes Required for Transport, HeLa Cells, Humans, Transcription Factors chemistry, Viral Nonstructural Proteins genetics, Bluetongue virus physiology, DNA-Binding Proteins physiology, Transcription Factors physiology, Viral Nonstructural Proteins physiology, Virus Assembly
- Abstract
The release of Bluetongue virus (BTV) and other members of the Orbivirus genus from infected host cells occurs predominantly by cell lysis, and in some cases, by budding from the plasma membrane. Two nonstructural proteins, NS3 and NS3A, have been implicated in this process. Here we show that both proteins bind to human Tsg101 and its ortholog from Drosophila melanogaster with similar strengths in vitro. This interaction is mediated by a conserved PSAP motif in NS3 and appears to play a role in virus release. The depletion of Tsg101 with small interfering RNA inhibits the release of BTV and African horse sickness virus, a related orbivirus, from HeLa cells up to fivefold and threefold, respectively. Like most other viral proteins which recruit Tsg101, NS3 also harbors a PPXY late-domain motif that allows NS3 to bind NEDD4-like ubiquitin ligases in vitro. However, the late-domain motifs in NS3 do not function as effectively in facilitating the release of mini Gag virus-like particles from 293T cells as the late domains from human immunodeficiency virus type 1, human T-cell leukemia virus, and Ebola virus. A mutagenesis study showed that the arginine residue in the PPRY motif is responsible for the low activity of the NS3 late-domain motifs. Our data suggest that the BTV late-domain motifs either recruit an antagonist that interferes with budding or fail to recruit an agonist which is different from NEDD4.
- Published
- 2006
- Full Text
- View/download PDF
47. Induction of human immunodeficiency virus type 1-specific T cells by a bluetongue virus tubule-vectored vaccine prime-recombinant modified virus Ankara boost regimen.
- Author
-
Larke N, Murphy A, Wirblich C, Teoh D, Estcourt MJ, McMichael AJ, Roy P, and Hanke T
- Subjects
- Animals, Bluetongue virus genetics, Bluetongue virus immunology, HIV Antibodies biosynthesis, Humans, Mice, Mice, Inbred BALB C, T-Lymphocyte Subsets immunology, Vaccines, DNA administration & dosage, Vaccines, DNA immunology, Vaccines, Synthetic administration & dosage, Viral Vaccines administration & dosage, Immunization, Secondary, T-Lymphocyte Subsets physiology, Vaccines, Synthetic immunology, Viral Vaccines immunology
- Abstract
In the absence of strategies for reliable induction of antibodies broadly neutralizing human immunodeficiency virus type 1 (HIV-1), vaccine efforts have shifted toward the induction of cell-mediated immunity. Here we describe the construction and immunogenicity of novel T-cell vaccine NS1.HIVA, which delivers the HIV-1 clade A consensus-derived immunogen HIVA on the surface of tubular structures spontaneously formed by protein NS1 of bluetongue virus. We demonstrated that NS1 tubules can accommodate a protein as large as 527 amino acids without losing their self-assembly capability. When injected into BALB/c mice by several routes, chimeric NS1.HIVA tubules induced HIV-1-specific major histocompatibility complex class I-restricted T cells. These could be boosted by modified virus Ankara expressing the same immunogen and generate a memory capable of gamma interferon (IFN-gamma) production, proliferation, and lysis of sensitized target cells. Induced memory T cells readily produced IFN-gamma 230 days postimmunization, and upon a surrogate virus challenge, NS1.HIVA vaccine alone decreased the vaccinia virus vv.HIVA load in ovaries by 2 orders of magnitude 280 days after immunization. Thus, because of its T-cell immunogenicity and antigenic simplicity, the NS1 delivery system could serve as a priming agent for heterologous prime-boost vaccination regimens. Its usefulness in primates, including humans, remains to be determined.
- Published
- 2005
- Full Text
- View/download PDF
48. A capsid protein of nonenveloped Bluetongue virus exhibits membrane fusion activity.
- Author
-
Forzan M, Wirblich C, and Roy P
- Subjects
- Amino Acid Motifs, Animals, Bluetongue virus chemistry, Bluetongue virus genetics, Capsid Proteins chemistry, Capsid Proteins genetics, Cell Fusion, Cell Line, Giant Cells cytology, Giant Cells virology, Receptors, Virus metabolism, Sequence Deletion genetics, Spodoptera, Bluetongue virus metabolism, Capsid Proteins metabolism, Cell Membrane metabolism, Membrane Fusion
- Abstract
The outer capsid layer of Bluetongue virus, a member of the nonenveloped Reoviridae family, is composed of two proteins, a receptor-binding protein, VP2, and a second protein, VP5, which shares structural features with class I fusion proteins of enveloped viruses. In the replication cycle of Bluetongue virus VP5 acts as a membrane permeabilization protein that mediates release of viral particles from endosomal compartments into the cytoplasm. Here, we show that VP5 can also act as a fusion protein and induce syncytium formation when it is fused to a transmembrane anchor and expressed on the cell surface. Fusion activity is strictly pH-dependent and is triggered by short exposure to low pH. No cell-cell fusion is observed at neutral pH. Deletion of the first 40 amino acids, which can fold into two amphipathic helices, abolishes fusion activity. Syncytium formation by VP5 is inhibited in the presence of VP2 when it is expressed in a membrane-anchored form. The data indicate an interaction between the outer capsid protein VP2 and VP5 and show that VP5 undergoes pH-dependent conformational changes that render it capable of interacting with cellular membranes. More importantly, our data show that a membrane permeabilization protein of a nonenveloped virus can evolve into a fusion protein by the addition of an appropriate transmembrane anchor. The results strongly suggest that the mechanism of membrane permeabilization by VP5 and membrane fusion by viral fusion proteins require similar structural features and conformational changes.
- Published
- 2004
- Full Text
- View/download PDF
49. Sequence specificity in the interaction of Bluetongue virus non-structural protein 2 (NS2) with viral RNA.
- Author
-
Lymperopoulos K, Wirblich C, Brierley I, and Roy P
- Subjects
- Animals, Bluetongue virus genetics, Cell Line, Nucleic Acid Conformation, RNA, Messenger chemistry, RNA, Messenger genetics, RNA, Messenger metabolism, RNA, Viral chemistry, RNA, Viral genetics, Viral Nonstructural Proteins chemistry, Bluetongue virus metabolism, RNA, Viral metabolism, Viral Nonstructural Proteins metabolism
- Abstract
The non-structural protein NS2 of Bluetongue virus (BTV) is synthesized abundantly in virus-infected cells and has been suggested to be involved in virus replication. The protein, with a high content of charged residues, possesses a strong affinity for single-stranded RNA species but, to date, all studies have failed to identify any specificity in the NS2-RNA interaction. In this report, we have examined, through RNA binding assays using highly purified NS2, the specificity of interaction with different single-stranded RNA (ssRNA) species in the presence of appropriate competitors. The data obtained show that NS2 indeed has a preference for BTV ssRNA over nonspecific RNA species and that NS2 recognizes a specific region within the BTV10 segment S10. The secondary structure of this region was determined and found to be a hairpin-loop with substructures within the loop. Modification-inhibition experiments highlighted two regions within this structure that were protected from ribonuclease cleavage in the presence of NS2. Overall, these data imply that a function of NS2 may be to recruit virus messenger RNAs (that also act as templates for synthesis of genomic RNAs) selectively from other RNA species within the infected cytosol of the cell during virus replication.
- Published
- 2003
- Full Text
- View/download PDF
50. Specific treatment of autoimmunity with recombinant invariant chains in which CLIP is replaced by self-epitopes.
- Author
-
Bischof F, Wienhold W, Wirblich C, Malcherek G, Zevering O, Kruisbeek AM, and Melms A
- Subjects
- Animals, Antigen Presentation immunology, Antigens, Differentiation, B-Lymphocyte genetics, Autoantigens genetics, Epitopes, T-Lymphocyte genetics, Female, Histocompatibility Antigens Class II genetics, Injections, Intravenous, Lymphocyte Activation immunology, Mice, Myelin Basic Protein genetics, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins immunology, Self Tolerance immunology, Antigens, Differentiation, B-Lymphocyte immunology, Autoantigens immunology, Autoimmunity immunology, Encephalomyelitis, Autoimmune, Experimental immunology, Epitopes, T-Lymphocyte immunology, Histocompatibility Antigens Class II immunology, Myelin Basic Protein immunology
- Abstract
The invariant chain (Ii) binds to newly synthesized MHC class II molecules with the CLIP region of Ii occupying the peptide-binding groove. Here we demonstrate that recombinant Ii proteins with the CLIP region replaced by antigenic self-epitopes are highly efficient in activating and silencing specific T cells in vitro and in vivo. The Ii proteins require endogenous processing by antigen-presenting cells for efficient T cell activation. An Ii protein encompassing the epitope myelin basic protein amino acids 84-96 (Ii-MBP84-96) induced the model autoimmune disease experimental allergic encephalomyelitis (EAE) with a higher severity and earlier onset than the peptide. When applied in a tolerogenic manner, Ii-MBP84-96 abolished antigen-specific T cell proliferation and suppressed peptide-induced EAE more effectively than peptide alone. Importantly, i.v. administration of Ii proteins after EAE induction completely abrogated the disease, whereas peptides only marginally suppressed disease symptoms. Ii fusion proteins are thus more efficient than peptide in modulating CD4(+) T cell-mediated autoimmunity, documenting their superior qualities for therapeutic antigen delivery in vivo.
- Published
- 2001
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.