Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 199
Filter
1.
Expert Rev Vaccines ; 23(1): 294-302, 2024.
Article in English | MEDLINE | ID: mdl-38372241

ABSTRACT

INTRODUCTION: Transmissible vaccines offer a novel approach to suppressing viruses in wildlife populations, with possible applications against viruses that infect humans as zoonoses - Lassa, Ebola, rabies. To ensure safety, current designs propose a recombinant vector platform in which the vector is isolated from the target wildlife population. Because using an endemic vector creates the potential for preexisting immunity to block vaccine transmission, these designs focus on vector viruses capable of superinfection, spreading throughout the host population following vaccination of few individuals. AREAS COVERED: We present original theoretical arguments that, regardless of its R0 value, a recombinant vaccine using a superinfecting vector is not expected to expand its active infection coverage when released into a wildlife population that already carries the vector. However, if superinfection occurs at a high rate such that individuals are repeatedly infected throughout their lives, the immunity footprint in the population can be high despite a low incidence of active vaccine infections. Yet we provide reasons that the above expectation is optimistic. EXPERT OPINION: High vaccine coverage will typically require repeated releases or release into a population lacking the vector, but careful attention to vector choice and vaccine engineering should also help improve transmissible vaccine utility.


Subject(s)
Rabies Vaccines , Rabies , Superinfection , Viruses , Humans , Animals , Rabies/prevention & control , Zoonoses/prevention & control , Rabies Vaccines/genetics , Vaccines, Synthetic/genetics
2.
Emerg Microbes Infect ; 13(1): 2300461, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38164714

ABSTRACT

During the COVID-19 epidemic, the incidence of rabies has increased in several countries, especially in remote and disadvantaged areas, due to inadequate surveillance and declining immunization coverage. Multiple vaccinations with inactivated rabies virus vaccines for pre- or post-exposure prophylaxis are considered inefficient, expensive and impractical in developing countries. Herein, three modified human recombinant adenoviruses type 5 designated Adv-RVG, Adv-E1-RVG, and Adv-RVDG, carrying rabies virus G (RVG) expression cassettes in various combinations within E1 or E3 genomic regions, were constructed to serve as rabies vaccine candidates. Adv-RVDG mediated greater RVG expression both in vitro and in vivo and induced a more robust and durable humoral immune response than the rabies vaccine strain SAD-L16, Adv-RVG, and Adv-E1-RVG by more effectively activating the dendritic cells (DCs) - follicular helper T (Tfh) cells - germinal centre (GC) / memory B cells (MBCs) - long-lived plasma cells (LLPCs) axis with 100% survival after a lethal RABV challenge in mice during the 24-week study period. Similarly, dogs and cats immunized with Adv-RVDG showed stronger and longer-lasting antibody responses than those vaccinated with a commercial inactivated rabies vaccine and showed good tolerance to Adv-RVDG. In conclusion, our study demonstrated that simultaneous insertion of protective antigens into the E1 and E3 genomic regions of adenovirus vector can significantly enhance the immunogenicity of adenoviral-vectored vaccines, providing a theoretical and practical basis for the subsequent development of multivalent and multi-conjugated vaccines using recombinant adenovirus platform. Meanwhile, our data suggest Adv-RVDG is a safe, efficient, and economical vaccine for mass-coverage immunization.


Subject(s)
Cat Diseases , Dog Diseases , Rabies Vaccines , Rabies virus , Cats , Dogs , Humans , Animals , Mice , Rabies virus/genetics , Rabies Vaccines/genetics , Immunity, Humoral , Antibodies, Viral , Adenoviridae/genetics
3.
Vaccine ; 42(5): 1116-1121, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38262810

ABSTRACT

Rabies is a lethal zoonotic disease that kills approximately 60,000 people each year. As the sole virion-surface protein, the rabies virus glycoprotein (RABV-G) mediates its host-cell entry. RABV-G's pre-fusion conformation displays major known neutralizing antibody epitopes, which can be used as immunogen for prophylaxis. H270P targeted mutation can stabilize RABV-G in the pre-fusion conformation. Herein, we report the development of a highly promising rabies mRNA vaccine composed of H270P targeted mutation packaged in lipid nanoparticle (LNP), named LNP-mRNA-G-H270P. Humoral and cellular immunity of this vaccine were assessed in mice comparing to the unmodified LNP-mRNA-G and a commercially available inactivated vaccine using one-way analysis of variance (ANOVA) followed by Dunnett's multiple comparisons test. The results show the titer of RABV-G-specific IgG and virus-neutralization antibody titers (VNTs) in LNP-mRNA-G-H270P group were significant higher than those in LNP-mRNA-G and inactivated vaccine groups. Likewise, IFN-γ-secreting splenocytes, level of IL-2 in the supernatant of spleen cells, as well as IFN-γ-producing CD4+ T cells in LNP-mRNA-G-H270P group were significant higher than those in the other two vaccine groups. Hence, these results demonstrated that targeting the H270P mutation in RABV-G through an mRNA-LNP vaccine platform represents a promising strategy for developing a more efficacious rabies vaccine.


Subject(s)
Rabies Vaccines , Rabies virus , Rabies , Humans , Animals , Mice , Rabies Vaccines/genetics , mRNA Vaccines , Immunity, Humoral , RNA, Messenger , Antibodies, Viral , Glycoproteins , Vaccines, Inactivated
4.
Int J Biol Macromol ; 255: 128085, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37977454

ABSTRACT

Rabies has been with humans for a long time, and its special transmission route and almost 100 % lethality rate made it once a nightmare for humans. In this study, by predicting the rabies virus glycoprotein outer membrane region and nucleoprotein B-cell antigenic epitopes, the coding sequence of the predicted highly antigenic polypeptide region obtained was assembled using the eukaryotic expression vector pcDNA3.1(-), and then E. coli was used as the delivery vector. The immunogenicity and protective properties of the vaccine were verified by in vivo and in vitro experiments, which demonstrated that the vaccine could produce antibodies in mice and prolong the survival time of mice exposed to the strong virus without any side effects. This study demonstrated that the preparation of an oral rabies DNA vaccine using food-borne microorganisms as a transport vehicle is feasible and could be a new strategy to eradicate rabies starting with wild animals.


Subject(s)
Rabies Vaccines , Rabies virus , Rabies , Vaccines, DNA , Humans , Animals , Mice , Rabies/prevention & control , Escherichia coli , Antibodies, Viral , Rabies Vaccines/genetics , Rabies virus/genetics , Epitopes, B-Lymphocyte/genetics
5.
Front Immunol ; 14: 1288879, 2023.
Article in English | MEDLINE | ID: mdl-37954577

ABSTRACT

Introduction: Rabies is a serious public health problem worldwide for which an effective treatment method is lacking but can be prevented by vaccines. Current vaccines are produced in cell or egg cultures, which are both costly and time consuming. Methods: Here, a non-replicating mRNA vaccine (RV021) encoding the rabies virus glycoprotein was developed in vitro, and its immunogenicity and protective efficacy against live virus was evaluated in mice. Results: A two-dose vaccination with 1 µg of RV021 at 7-day intervals induced a protective level of neutralizing antibody that was maintained for at least 260 days. RV021 induced a robust cellular immune response that was significantly superior to that of an inactivated vaccine. Two doses of 1 µg RV021 provided full protection against challenge with CVS of 30~60-fold lethal dose, 50%. Vaccine potency testing (according to the National Institutes of Health) in vivo revealed that the potency of RV021 at 15 µg/dose was 7.5 IU/dose, which is substantially higher than the standard for lot release of rabies vaccines for current human use. Conclusion: The mRNA vaccine RV021 induces a strong protective immune response in mice, providing a new and promising strategy for human rabies prevention and control.


Subject(s)
Rabies Vaccines , Rabies virus , Rabies , United States , Animals , Humans , Mice , Rabies/prevention & control , Rabies Vaccines/genetics , Antibodies, Viral , Antibodies, Neutralizing , Rabies virus/genetics
6.
Microb Pathog ; 185: 106425, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37923181

ABSTRACT

Rabies, caused by the rabies virus (RABV), is the most fatal zoonotic disease. It is a neglected tropical disease which remains a major public health problem, causing approximately 59,000 deaths worldwide annually. Despite the existence of effective vaccines, the high incidence of human rabies is mainly linked to tedious vaccine immunisation procedures and the overall high cost of post-exposure prophylaxis. Therefore, it is necessary to develop an effective vaccine that has a simple procedure and is affordable to prevent rabies infection in humans. RABV belongs to the genus Lyssavirus and family Rhabdoviridae. Previous phylogenetic analyses have identified seven major clades of RABV in China (China I-VII), confirmed by analysing nucleotide sequences from both the G and N proteins. This study evaluated the immunogenicity and protective capacity of SYS6008, an mRNA rabies vaccine expressing rabies virus glycoprotein, in mice and cynomolgus macaques. We demonstrated that SYS6008 induced sufficient levels of rabies neutralising antibody (RVNA) in mice. In addition, SYS6008 elicited strong and durable RVNA responses in vaccinated cynomolgus macaques. In the pre-exposure prophylaxis murine model, one or two injections of SYS6008 at 1/10 or 1/30 of dosage provided protection against a challenge with a 30-fold LD50 of rabies virus (China I and II clades). We also demonstrated that in the post-exposure prophylaxis murine model, which was exposed to lethal rabies virus (China I-VII clades) before vaccination, one or two injections of SYS6008 at both 1/10 and 1/30 dosages provided better protection against rabies virus challenge than the immunization by five injections of commercial vaccines at the same dosage. In addition, we proved that SYS6008-induced RVNAs could neutralise RABV from the China I-VII clades. Finally, 1/10 of the dosage of SYS6008 was able to stimulate significant RABV-G specificity in the T cell response. Furthermore, we found that SYS6008 induced high cellular immunity, including RABV-G-specific T cell responses and memory B cells. Our results imply that the SYS6008 rabies vaccine, with a much simpler vaccination procedure, better immunogenicity, and enhanced protective capacity, could be a candidate vaccine for post-exposure prophylaxis of rabies infections.


Subject(s)
Rabies Vaccines , Rabies virus , Rabies , Humans , Animals , Mice , Rabies/prevention & control , Rabies Vaccines/genetics , Rabies virus/genetics , Post-Exposure Prophylaxis/methods , Disease Models, Animal , Phylogeny , Antibodies, Viral , Macaca
7.
Int J Med Sci ; 20(10): 1282-1292, 2023.
Article in English | MEDLINE | ID: mdl-37786447

ABSTRACT

Rabies continues to be a huge threat to public health. The rabies virus envelope glycoprotein (RABV G) is a major rabies virus antigen and contains neutralizing epitopes, which are primary candidates for subunit vaccines and diagnostic antigens. However, the production and purification of rRABV G while retaining its antigenic and immunogenic remains to be a challenge. Here, we aimed to establish a platform for rRABV G production and purification, and determine the immunogenicity and antigenicity of rRABV G. The cDNA fragment encoding the soluble form of RABV G was synthesized and cloned into a lentiviral expressing vector. Recombinant lentiviral vector LV-CMV-RABV G-eGFP was packaged, titered, and then transduced into HEK 293T cells. The cell culture supernatant was purified using nickel affinity chromatography and subsequently confirmed through Western Blot analysis and indirect enzyme-linked immunosorbent assay (ELISA). The ELISA utilized human sera obtained from individuals who had been vaccinated with the human commercial Purified Vero Cells Rabies Vaccine (PVRV). Notably, we observed a neutralizing antibody response in immunized pigs rather than in mice. This discrepancy could potentially be attributed to factors such as the instability of the rRABV G protein, variations in host responses, and variances in the adjuvant used. Taking all these findings into account, the rRABV G protein generated in this study exhibits promise as a potential vaccine candidate for the prevention of rabies.


Subject(s)
Rabies Vaccines , Rabies virus , Rabies , Chlorocebus aethiops , Humans , Animals , Mice , Swine , Rabies virus/genetics , Rabies/prevention & control , HEK293 Cells , Vero Cells , Antibodies, Viral , Glycoproteins/genetics , Rabies Vaccines/genetics , Viral Envelope Proteins/genetics , Recombinant Proteins
8.
Nat Commun ; 14(1): 3713, 2023 06 22.
Article in English | MEDLINE | ID: mdl-37349310

ABSTRACT

Licensed rabies virus vaccines based on whole inactivated virus are effective in humans. However, there is a lack of detailed investigations of the elicited immune response, and whether responses can be improved using novel vaccine platforms. Here we show that two doses of a lipid nanoparticle-formulated unmodified mRNA vaccine encoding the rabies virus glycoprotein (RABV-G) induces higher levels of RABV-G specific plasmablasts and T cells in blood, and plasma cells in the bone marrow compared to two doses of Rabipur in non-human primates. The mRNA vaccine also generates higher RABV-G binding and neutralizing antibody titers than Rabipur, while the degree of somatic hypermutation and clonal diversity of the response are similar for the two vaccines. The higher overall antibody titers induced by the mRNA vaccine translates into improved cross-neutralization of related lyssavirus strains, suggesting that this platform has potential for the development of a broadly protective vaccine against these viruses.


Subject(s)
Rabies Vaccines , Rabies virus , Rabies , Animals , Humans , Rabies/prevention & control , Rabies Vaccines/genetics , Broadly Neutralizing Antibodies , RNA, Messenger , Antibodies, Viral , Rabies virus/genetics , Glycoproteins
9.
Elife ; 122023 05 25.
Article in English | MEDLINE | ID: mdl-37227428

ABSTRACT

Background: Dog-mediated rabies is endemic across Africa causing thousands of human deaths annually. A One Health approach to rabies is advocated, comprising emergency post-exposure vaccination of bite victims and mass dog vaccination to break the transmission cycle. However, the impacts and cost-effectiveness of these components are difficult to disentangle. Methods: We combined contact tracing with whole-genome sequencing to track rabies transmission in the animal reservoir and spillover risk to humans from 2010 to 2020, investigating how the components of a One Health approach reduced the disease burden and eliminated rabies from Pemba Island, Tanzania. With the resulting high-resolution spatiotemporal and genomic data, we inferred transmission chains and estimated case detection. Using a decision tree model, we quantified the public health burden and evaluated the impact and cost-effectiveness of interventions over a 10-year time horizon. Results: We resolved five transmission chains co-circulating on Pemba from 2010 that were all eliminated by May 2014. During this period, rabid dogs, human rabies exposures and deaths all progressively declined following initiation and improved implementation of annual islandwide dog vaccination. We identified two introductions to Pemba in late 2016 that seeded re-emergence after dog vaccination had lapsed. The ensuing outbreak was eliminated in October 2018 through reinstated islandwide dog vaccination. While post-exposure vaccines were projected to be highly cost-effective ($256 per death averted), only dog vaccination interrupts transmission. A combined One Health approach of routine annual dog vaccination together with free post-exposure vaccines for bite victims, rapidly eliminates rabies, is highly cost-effective ($1657 per death averted) and by maintaining rabies freedom prevents over 30 families from suffering traumatic rabid dog bites annually on Pemba island. Conclusions: A One Health approach underpinned by dog vaccination is an efficient, cost-effective, equitable, and feasible approach to rabies elimination, but needs scaling up across connected populations to sustain the benefits of elimination, as seen on Pemba, and for similar progress to be achieved elsewhere. Funding: Wellcome [207569/Z/17/Z, 095787/Z/11/Z, 103270/Z/13/Z], the UBS Optimus Foundation, the Department of Health and Human Services of the National Institutes of Health [R01AI141712] and the DELTAS Africa Initiative [Afrique One-ASPIRE/DEL-15-008] comprising a donor consortium of the African Academy of Sciences (AAS), Alliance for Accelerating Excellence in Science in Africa (AESA), the New Partnership for Africa's Development Planning and Coordinating (NEPAD) Agency, Wellcome [107753/A/15/Z], Royal Society of Tropical Medicine and Hygiene Small Grant 2017 [GR000892] and the UK government. The rabies elimination demonstration project from 2010-2015 was supported by the Bill & Melinda Gates Foundation [OPP49679]. Whole-genome sequencing was partially supported from APHA by funding from the UK Department for Environment, Food and Rural Affairs (Defra), Scottish government and Welsh government under projects SEV3500 and SE0421.


Subject(s)
Bites and Stings , Dog Diseases , Rabies Vaccines , Rabies , Dogs , Animals , Humans , Rabies/epidemiology , Rabies/prevention & control , Rabies/veterinary , Contact Tracing , Cost-Benefit Analysis , Rabies Vaccines/genetics , Tanzania/epidemiology , Genomics , Bites and Stings/epidemiology , Dog Diseases/epidemiology , Dog Diseases/prevention & control
10.
Proc Natl Acad Sci U S A ; 120(11): e2216667120, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36877838

ABSTRACT

Transmissible vaccines are an emerging biotechnology that hold prospects to eliminate pathogens from wildlife populations. Such vaccines would genetically modify naturally occurring, nonpathogenic viruses ("viral vectors") to express pathogen antigens while retaining their capacity to transmit. The epidemiology of candidate viral vectors within the target wildlife population has been notoriously challenging to resolve but underpins the selection of effective vectors prior to major investments in vaccine development. Here, we used spatiotemporally replicated deep sequencing to parameterize competing epidemiological mechanistic models of Desmodus rotundus betaherpesvirus (DrBHV), a proposed vector for a transmissible vaccine targeting vampire bat-transmitted rabies. Using 36 strain- and location-specific time series of prevalence collected over 6 y, we found that lifelong infections with cycles of latency and reactivation, combined with a high R0 (6.9; CI: 4.39 to 7.85), are necessary to explain patterns of DrBHV infection observed in wild bats. These epidemiological properties suggest that DrBHV may be suited to vector a lifelong, self-boosting, and transmissible vaccine. Simulations showed that inoculating a single bat with a DrBHV-vectored rabies vaccine could immunize >80% of a bat population, reducing the size, frequency, and duration of rabies outbreaks by 50 to 95%. Gradual loss of infectious vaccine from vaccinated individuals is expected but can be countered by inoculating larger but practically achievable proportions of bat populations. Parameterizing epidemiological models using accessible genomic data brings transmissible vaccines one step closer to implementation.


Subject(s)
Betaherpesvirinae , Chiroptera , Rabies Vaccines , Rabies , Humans , Animals , Rabies Vaccines/genetics , Rabies/epidemiology , Rabies/prevention & control , Rabies/veterinary , Vaccination/veterinary , Animals, Wild
11.
Adv Exp Med Biol ; 1407: 191-208, 2023.
Article in English | MEDLINE | ID: mdl-36920698

ABSTRACT

Lyssaviruses, which belong to the family Rhabdoviridae, are enveloped and bullet-shaped ssRNA viruses with genetic diversity. All members of Lyssavirus genus are known to infect warm-blooded animals and cause the fatal disease rabies. The rabies virus (RABV) in lyssavirus is the major pathogen to cause fatal rabies. The pseudotyped RABV is constructed to study the biological functions of G protein and evaluation of anti-RABV products including vaccine-induced antisera, rabies immunoglobulins (RIG), neutralizing mAbs, and other antiviral inhibitors. In this chapter, we focus on RABV as a representative and describe the construction of RABV G protein bearing pseudotyped virus and its applications. Other non-RABV lyssaviruses are also included.


Subject(s)
Lyssavirus , Rabies Vaccines , Rabies virus , Rabies , Rhabdoviridae Infections , Animals , Lyssavirus/genetics , Viral Pseudotyping , Rabies virus/genetics , Rabies Vaccines/genetics , Rabies Vaccines/metabolism
12.
Microbiol Immunol ; 67(2): 69-78, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36346082

ABSTRACT

Rabies is a viral disease that is nearly 100% fatal once clinical signs and symptoms develop. Post-exposure prophylaxis can efficiently prevent rabies, and antibody (Ab) induction by vaccination or passive immunization of human rabies immunoglobulin (HRIG) or monoclonal antibodies (mAbs) play an integral role in prevention against rabies. In addition to their capacity to neutralize viruses, antibodies exert their antiviral effects by antibody-dependent cellular cytotoxicity (ADCC), which plays an important role in antiviral immunity and clearance of viral infections. For antibodies against rabies virus (RABV), evaluation of ADCC activity was neglected. Here, we developed a robust cell-based reporter gene assay (RGA) for the determination of the ADCC activity of anti-RABV antibodies using CVS-N2c-293 cells, which stably express the glycoprotein (G) of RABV strain CVS-N2c as target cells, and Jurkat cells, which stably express FcγRⅢa and nuclear factor of activated T cells (NFAT) reporter gene as effector cells (Jurkat/NFAT-luc/FcγRⅢa cells). The experimental parameters were carefully optimized, and the established ADCC assay was systematically validated according to the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) Q2 guideline. We also evaluated the ADCC activity of anti-RABV antibodies, including mAbs, HRIG, and vaccine induced antisera, and found that all test antibodies exhibited ADCC activity with varied strengths. The established RGA provides a novel method for evaluating the ADCC of anti-RABV antibodies.


Subject(s)
Rabies Vaccines , Rabies , Humans , Antibodies, Viral , Genes, Reporter , Rabies Vaccines/genetics , Antibody-Dependent Cell Cytotoxicity , Antibodies, Monoclonal , Glycoproteins/genetics , Antiviral Agents
13.
Virol J ; 19(1): 184, 2022 11 12.
Article in English | MEDLINE | ID: mdl-36371169

ABSTRACT

Rabies is a lethal zoonotic disease that is mainly caused by the rabies virus (RABV). Although effective vaccines have long existed, current vaccines take both time and cost to produce. Messenger RNA (mRNA) technology is an emergent vaccine platform that supports rapid vaccine development on a large scale. Here, an optimized mRNA vaccine construct (LVRNA001) expressing rabies virus glycoprotein (RABV-G) was developed in vitro and then evaluated in vivo for its immunogenicity and protective capacity in mice and dogs. LVRNA001 induced neutralizing antibody production and a strong Th1 cellular immune response in mice. In both mice and dogs, LVRNA001 provided protection against challenge with 50-fold lethal dose 50 (LD50) of RABV. With regards to protective efficiency, an extended dosing interval (14 days) induced greater antibody production than 3- or 7-day intervals in mice. Finally, post-exposure immunization against RABV was performed to evaluate the survival rates of dogs receiving two 25 µg doses of LVRNA001 vs. five doses of inactivated vaccine over the course of three months. Survival rate in the LVRNA001 group was 100%, whereas survival rate in the inactivated vaccine control group was only 33.33%. In conclusion, these results demonstrated that LVRNA001 induced strong protective immune responses in mice and dogs, which provides a new and promising prophylactic strategy for rabies.


Subject(s)
Rabies Vaccines , Rabies virus , Rabies , Dogs , Mice , Animals , Rabies Vaccines/genetics , RNA, Messenger , Antibodies, Viral , Rabies virus/genetics , Vaccines, Inactivated , Antibody Formation , mRNA Vaccines
14.
Virulence ; 13(1): 1446-1454, 2022 12.
Article in English | MEDLINE | ID: mdl-35999776

ABSTRACT

Rabies is an important zoonotic disease caused by the rabies virus (RABV). Currently, no effective treatment is available for this condition. The prevention and control of rabies mainly depend on effective vaccination. Therefore, it is crucial to enhance the immune responses induced by the rabies vaccine. Virus neutralizing antibodies (VNA) induced by rabies vaccines are important for the clearance of RABV. Interleukin-25 (IL-25) has been demonstrated to activate T helper type 2 cells that contribute to humoral immune responses. The IL-25 gene was inserted into the genome of RABV, and the immunogenicity of recombinant RABV with IL-25 gene was investigated to develop more efficient rabies vaccines. Here, we found that the expression of IL-25 did not affect RABV production in vitro and pathogenicity in vivo. However, recombinant RABV expression of IL-25 induced a better VNA level than the parental virus in mice. In addition, expression of IL-25 enhanced the IgG1 level induced by RABV. Furthermore, mice immunized with recombinant RABV showed a higher survival rate and milder clinical signs than those immunized with the parent strain after challenge with CVS-11. Thus, these results showed that IL-25 could enhance the humoral immune responses induced by RABV, suggesting that IL-25 can be used as a viral vaccine adjuvant.


Subject(s)
Rabies Vaccines , Rabies virus , Rabies , Animals , Antibodies, Viral , Immunity, Humoral , Interleukin-17/genetics , Mice , Rabies/prevention & control , Rabies Vaccines/genetics , Rabies virus/genetics
15.
Cell Host Microbe ; 30(9): 1219-1230.e7, 2022 09 14.
Article in English | MEDLINE | ID: mdl-35985336

ABSTRACT

Rabies virus (RABV) causes lethal encephalitis and is responsible for approximately 60,000 deaths per year. As the sole virion-surface protein, the rabies virus glycoprotein (RABV-G) mediates host-cell entry. RABV-G's pre-fusion trimeric conformation displays epitopes bound by protective neutralizing antibodies that can be induced by vaccination or passively administered for post-exposure prophylaxis. We report a 2.8-Å structure of a RABV-G trimer in the pre-fusion conformation, in complex with two neutralizing and protective monoclonal antibodies, 17C7 and 1112-1, that recognize distinct epitopes. One of these antibodies is a licensed prophylactic (17C7, Rabishield), which we show locks the protein in pre-fusion conformation. Targeted mutations can similarly stabilize RABV-G in the pre-fusion conformation, a key step toward structure-guided vaccine design. These data reveal the higher-order architecture of a key therapeutic target and the structural basis of neutralization by antibodies binding two key antigenic sites, and this will facilitate the development of improved vaccines and prophylactic antibodies.


Subject(s)
Rabies Vaccines , Rabies virus , Rabies , Antibodies, Monoclonal , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral , Epitopes , Glycoproteins/genetics , Humans , Membrane Proteins , Rabies/drug therapy , Rabies/prevention & control , Rabies Vaccines/genetics
16.
Front Immunol ; 13: 1099991, 2022.
Article in English | MEDLINE | ID: mdl-36761167

ABSTRACT

Background: Rabies is a lethal zoonotic disease that kills approximately 60,000 people each year. Although inactivated rabies vaccines are available, multiple-dose regimensare recommended for pre-exposure prophylaxis or post-exposure prophylaxis,which cuts down the cost- and time-effectiveness, especially in low- and middle incomecountries. Methods: We developed a nucleoside-modified Rabies mRNA-lipid nanoparticle vaccine (RABV-G mRNA-LNP) encoding codon-optimized viral glycoprotein and assessed the immunogenicity and protective efficacy of this vaccine in mice comparing to a commercially available inactivated vaccine. Results: We first showed that, when evaluated in mice, a single vaccination of RABV-G mRNA with a moderate or high dose induces more potent humoral and T-cell immune responses than that elicited by three inoculations of the inactivated vaccine. Importantly, mice receiving a single immunization of RABV-G mRNA, even at low doses, showed full protection against the lethal rabies challenge. We further demonstrated that the humoral immune response induced by single RABV-G mRNA vaccination in mice could last for at least 25 weeks, while a two-dose strategy could extend the duration of the highly protective response to one year or even longer. In contrast, the three-dose regimen of inactivated vaccine failed to do so. Conclusion: Our study confirmed that it is worth developing a single-dose nucleoside-modified Rabies mRNA-LNP vaccine, which could confer much prolonged and more effective protection.


Subject(s)
Rabies Vaccines , Rabies , Animals , Mice , Rabies Vaccines/genetics , Rabies/prevention & control , Nucleosides , RNA, Messenger/genetics , Antibodies, Viral , Vaccination , Immunity, Humoral , Vaccines, Inactivated
17.
Viruses ; 13(11)2021 11 16.
Article in English | MEDLINE | ID: mdl-34835093

ABSTRACT

Rabies is a lethal zoonotic disease caused by lyssaviruses, such as rabies virus (RABV), that results in nearly 100% mortality once clinical symptoms appear. There are no curable drugs available yet. RABV contains five structural proteins that play an important role in viral replication, transcription, infection, and immune escape mechanisms. In the past decade, progress has been made in research on the pathogenicity of RABV, which plays an important role in the creation of new recombinant RABV vaccines by reverse genetic manipulation. Here, we review the latest advances on the interaction between RABV proteins in the infected host and the applied development of rabies vaccines by using a fully operational RABV reverse genetics system. This article provides a background for more in-depth research on the pathogenic mechanism of RABV and the development of therapeutic drugs and new biologics.


Subject(s)
Rabies Vaccines/immunology , Rabies virus/immunology , Rabies/prevention & control , Viral Structural Proteins/immunology , Animals , Humans , Rabies/immunology , Rabies/virology , Rabies Vaccines/genetics , Rabies virus/genetics , Reverse Genetics/methods , Vaccines, Attenuated , Viral Structural Proteins/genetics , Virus Replication
18.
Mol Biotechnol ; 63(11): 1068-1080, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34228257

ABSTRACT

Rabies is an ancient zoonotic disease that still causes the death of over 59,000 people worldwide each year. The rabies lyssavirus encodes five proteins, including the envelope glycoprotein and the matrix protein. RVGP is the only protein exposed on the surface of viral particle, and it can induce immune response with neutralizing antibody formation. RVM has the ability to assist with production process of virus-like particles. VLPs were produced in recombinant baculovirus system. In this work, two recombinant baculoviruses carrying the RVGP and RVM genes were constructed. From the infection and coinfection assays, we standardized the best multiplicity of infection and the best harvest time. Cell supernatants were collected, concentrated, and purified by sucrose gradient. Each step was used for protein detection through immunoassays. Sucrose gradient analysis enabled to verify the separation of VLPs from rBV. Through the negative contrast technique, we visualized structures resembling rabies VLPs produced in insect cells and rBV in the different fractions of the sucrose gradient. Using ELISA to measure total RVGP, the recovery efficiency of VLPs at each stage of the purification process was verified. Thus, these results encourage further studies to confirm whether rabies VLPs are a promising candidate for a veterinary rabies vaccine.


Subject(s)
Baculoviridae/genetics , Insecta/metabolism , Rabies Vaccines/biosynthesis , Rabies virus/metabolism , Rabies/virology , Vaccines, Virus-Like Particle/biosynthesis , Animals , Baculoviridae/isolation & purification , Baculoviridae/metabolism , Cells, Cultured , Humans , Insecta/immunology , Insecta/virology , Rabies Vaccines/genetics , Rabies Vaccines/immunology , Rabies Vaccines/isolation & purification , Rabies virus/immunology , Rabies virus/isolation & purification , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/isolation & purification
19.
Vaccine ; 39(28): 3777-3784, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34092430

ABSTRACT

To improve the safety of genetically modified live rabies vaccine strains, most studies have utilized an attenuating Arg-to-Glu mutation at position 333 in the glycoprotein (G333), which is responsible for attenuation of the live vaccine strain SAG2. The Glu residue requires two nucleotide substitutions to revert to pathogenic Arg, thus significantly lowering the probability of pathogenic reversion caused by the Glu-to-Arg mutation at G333. However, only one nucleotide substitution is sufficient to convert the Glu residue to another pathogenic residue, Lys, and thereby to cause pathogenic reversion. This indicates a potential safety problem of SAG2 and the live vaccine candidates attenuated by Glu at G333. In this study, aiming to solve this problem, we examined the utility of a Leu residue, which requires two nucleotide substitutions to be both Arg and Lys, as an attenuating mutation at G333. Using a reverse genetics system of the live vaccine strain ERA, we generated ERA-G333Leu by introducing an Arg-to-Leu mutation at G333. Similar to ERA-G333Glu, which is attenuated by an Arg-to-Glu mutation at G333, ERA-G333Leu did not cause obvious clinical signs in 6-week-old mice after intracerebral inoculation. Importantly, after 10 passages in suckling mouse brains, ERA-G333Glu acquired a pathogenic Lys or Arg at G333 and a high level of lethality in mice, whereas ERA-G333Leu retained the attenuating Leu at G333 and only showed a modest level of virulence probably caused by a mutation at G194. In addition, ERA-G333Leu and ERA-G333Glu induced neutralizing antibody response and protective immunity in mice with similar efficiencies. The results demonstrate that, compared to ERA-G333Glu, ERA-G333Leu is more stably attenuated, also indicating the high utility of a Leu residue as an attenuating mutation at G333 in the development of live rabies vaccine strains with a high level of safety.


Subject(s)
Rabies Vaccines , Rabies virus , Rabies , Animals , Glycoproteins/genetics , Mice , Rabies/prevention & control , Rabies Vaccines/genetics , Vaccines, Attenuated/genetics
20.
Vet Microbiol ; 251: 108906, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33160196

ABSTRACT

Rabies remains an important public health threat in most developing countries. To develop a more effective and safe oral vaccine against rabies, we constructed recombinant Lactobacillus plantarum NC8 carrying one or two copies of the G gene with a dendritic cell-targeting peptide (DCpep) fused at the C-terminal designated NC8-pSIP409-sRVG or NC8-pSIP409-dRVG, respectively. The immunogenicity and protective efficacy of these recombinant Lactobacillus plantarum against RABV were evaluated by oral administration in a mouse model. The results showed that recombinant NC8-pSIP409-dRVG possessed more G protein, resulting in more functional maturation of DCs. After three cycle of oral immunization, NC8-pSIP409-dRVG induced significantly higher levels of specific IgG antibody and mixed Th1/Th2 with a strong Th1-biasd immune response in mice. Most importantly, although the titers of RABV neutralizing antibody (VNA) were below the threshold of 0.5 IU/mL, the NC8-pSIP409-dRVG could protect 60 % of inoculated mice against lethal RABV challenge. These data reveal that recombinant NC8-pSIP409-dRVG may be a novel and promising oral vaccine candidate to prevent and control of animal rabies.


Subject(s)
GTP-Binding Proteins/genetics , Lactobacillus plantarum/genetics , Peptides/genetics , Rabies Vaccines/genetics , Rabies Vaccines/immunology , Rabies/prevention & control , Rabies/veterinary , Administration, Oral , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Chickens/immunology , Dendritic Cells/immunology , Female , GTP-Binding Proteins/administration & dosage , GTP-Binding Proteins/immunology , Immunity, Cellular , Lactobacillus plantarum/immunology , Mice , Mice, Inbred BALB C , Peptides/administration & dosage , Peptides/immunology , Rabies Vaccines/administration & dosage , Rabies Vaccines/standards , Th1 Cells/immunology , Th2 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...