Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 203
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(11): e2216667120, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36877838

RESUMEN

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.


Asunto(s)
Betaherpesvirinae , Quirópteros , Vacunas Antirrábicas , Rabia , Humanos , Animales , Vacunas Antirrábicas/genética , Rabia/epidemiología , Rabia/prevención & control , Rabia/veterinaria , Vacunación/veterinaria , Animales Salvajes
2.
Virol J ; 21(1): 154, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38978059

RESUMEN

BACKGROUND: Rabies is a fatal zoonotic disease whose pathogenesis has not been fully elucidated, and vaccination is the only effective method for protecting against rabies virus infection. Most inactivated vaccines are produced using Vero cells, which are African green monkey kidney cells, to achieve large-scale production. However, there is a potential carcinogenic risk due to nonhuman DNA contamination. Thus, replacing Vero cells with human diploid cells may be a safer strategy. In this study, we developed a novel 2BS cell-adapted rabies virus strain and analysed its sequence, virulence and immunogenicity to determine its application potential as a human diploid cell inactivated vaccine. METHODS AND RESULTS: The 2BS cell-adapted rabies virus strain 2aG4-B40 was established by passage for 40 generations and selection of plaques in 2BS cells. RNA sequence analysis revealed that mutations in 2BS cell-adapted strains were not located at key sites that regulate the production of neutralizing antibodies or virulence in the aG strain (GQ412744.1). The gradual increase in virulence (remaining above 7.0 logLD50/ml from the 40th to 55th generation) and antigen further indicated that these mutations may increase the affinity of the adapted strains for human diploid cells. Identification tests revealed that the 2BS cell-adapted virus strain was neutralized by anti-rabies serum, with a neutralization index of 19,952. PrEP and PEP vaccination and the NIH test further indicated that the vaccine prepared with the 2aG4-B40 strain had high neutralizing antibody levels (2.24 to 46.67 IU/ml), immunogenicity (protection index 270) and potency (average 11.6 IU/ml). CONCLUSIONS: In this study, a 2BS cell-adapted strain of the 2aG4 rabies virus was obtained by passage for 40 generations. The results of sequencing analysis and titre determination of the adapted strain showed that the mutations in the adaptive process are not located at key sequence regions of the virus, and these mutations may enhance the affinity of the adapted strain for human diploid cells. Moreover, vaccines made from the adapted strain 2aG4-B40 had high potency and immunogenicity and could be an ideal candidate rabies virus strain for inactivated vaccine preparation.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Vacunas Antirrábicas , Virus de la Rabia , Rabia , Virus de la Rabia/inmunología , Virus de la Rabia/genética , Virus de la Rabia/patogenicidad , Animales , Vacunas Antirrábicas/inmunología , Vacunas Antirrábicas/genética , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/sangre , Rabia/prevención & control , Rabia/inmunología , Rabia/virología , Humanos , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Chlorocebus aethiops , Virulencia , Vacunas de Productos Inactivados/inmunología , Células Vero , China , Ratones , Línea Celular , Mutación , Femenino , Inmunogenicidad Vacunal
3.
Microb Pathog ; 185: 106425, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37923181

RESUMEN

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.


Asunto(s)
Vacunas Antirrábicas , Virus de la Rabia , Rabia , Humanos , Animales , Ratones , Rabia/prevención & control , Vacunas Antirrábicas/genética , Virus de la Rabia/genética , Profilaxis Posexposición/métodos , Modelos Animales de Enfermedad , Filogenia , Anticuerpos Antivirales , Macaca
4.
Microbiol Immunol ; 67(2): 69-78, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36346082

RESUMEN

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.


Asunto(s)
Vacunas Antirrábicas , Rabia , Humanos , Anticuerpos Antivirales , Genes Reporteros , Vacunas Antirrábicas/genética , Citotoxicidad Celular Dependiente de Anticuerpos , Anticuerpos Monoclonales , Glicoproteínas/genética , Antivirales
5.
Int J Med Sci ; 20(10): 1282-1292, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37786447

RESUMEN

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.


Asunto(s)
Vacunas Antirrábicas , Virus de la Rabia , Rabia , Chlorocebus aethiops , Humanos , Animales , Ratones , Porcinos , Virus de la Rabia/genética , Rabia/prevención & control , Células HEK293 , Células Vero , Anticuerpos Antivirales , Glicoproteínas/genética , Vacunas Antirrábicas/genética , Proteínas del Envoltorio Viral/genética , Proteínas Recombinantes
6.
Adv Exp Med Biol ; 1407: 191-208, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36920698

RESUMEN

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.


Asunto(s)
Lyssavirus , Vacunas Antirrábicas , Virus de la Rabia , Rabia , Infecciones por Rhabdoviridae , Animales , Lyssavirus/genética , Pseudotipado Viral , Virus de la Rabia/genética , Vacunas Antirrábicas/genética , Vacunas Antirrábicas/metabolismo
7.
Virol J ; 19(1): 184, 2022 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-36371169

RESUMEN

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.


Asunto(s)
Vacunas Antirrábicas , Virus de la Rabia , Rabia , Perros , Ratones , Animales , Vacunas Antirrábicas/genética , ARN Mensajero , Anticuerpos Antivirales , Virus de la Rabia/genética , Vacunas de Productos Inactivados , Formación de Anticuerpos , Vacunas de ARNm
8.
Protein Expr Purif ; 168: 105567, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31904423

RESUMEN

Rabies is a fatal zoonosis which could affect all mammals. Glycoprotein (G protein) from the rabies virus plays an important role in the binding of virus to target cells. However, expression of the G protein with native conformation has been a great challenge for many years. In this study, we solved this problem by replacing the original signal peptide of rabies virus G protein with the one from the heavy chain of human IgG. The expression levels of recombinant G protein dramatically increased from a few µg/L to 50 mg/L in the culture supernatants. The identity of the recombinant G protein was confirmed by western blotting using both 6XHis mAb 6E2 and rabies G protein mAb 7G3. The correct conformation of the recombinant G protein was shown by using rabies virus neutralizing antibodies. In addition, the recombinant G protein had immune-reactivities with mice sera raised against rabies vaccines and vice versa. Taken together, our data suggested that by replacing the signal peptide, the expression level of the G protein with native conformation could be significantly improved. This would help the development of a rabies subunit vaccine, structural studies of rabies G protein, elucidation of the signal pathway of RABV infection.


Asunto(s)
Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , Antígenos Virales/administración & dosificación , Cadenas Pesadas de Inmunoglobulina/genética , Virus de la Rabia/inmunología , Rabia/prevención & control , Proteínas Recombinantes de Fusión/genética , Proteínas del Envoltorio Viral/administración & dosificación , Animales , Antígenos Virales/genética , Antígenos Virales/inmunología , Clonación Molecular , Protección Cruzada , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Sueros Inmunes/química , Inmunoglobulina G/genética , Inmunoglobulina G/metabolismo , Cadenas Pesadas de Inmunoglobulina/metabolismo , Ratones , Ingeniería de Proteínas/métodos , Señales de Clasificación de Proteína/genética , Rabia/virología , Vacunas Antirrábicas/administración & dosificación , Vacunas Antirrábicas/biosíntesis , Vacunas Antirrábicas/genética , Virus de la Rabia/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
9.
J Infect Dis ; 220(9): 1521-1528, 2019 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-31374568

RESUMEN

BACKGROUND: Ebola virus (EBOV) is a highly lethal member of the Filoviridae family associated with human hemorrhagic disease. Despite being a sporadic disease, it caused a large outbreak in 2014-2016 in West Africa and another outbreak recently in the Democratic Republic of Congo. Several vaccine candidates are currently in preclinical and clinical studies but none are stable without cold chain storage. METHODS: We used preservation by vaporization (PBV), a novel processing technology to heat-stabilize FiloRab1 (inactivated rabies-based Ebola vaccine), a candidate Ebola vaccine, and stored the vials at temperatures ranging from 4°C to 50°C for 10 days to 12 months. We immunized Syrian hamsters with the best long-term stable FiloRab1 PBV vaccines and challenged them with rabies virus (RABV). RESULTS: Syrian hamsters immunized with FiloRab1 PBV-processed vaccines stored at temperatures of 4°C and 37°C for 6 months, and at 50°C for 2 weeks, seroconverted against both RABV-G and EBOV-GP. Notably, all of the FiloRab1 PBV vaccines proved to be 100% effective in a RABV challenge model. CONCLUSIONS: We successfully demonstrated that the FiloRab1 PBV vaccines are stable and efficacious for up to 6 months when stored at temperatures ranging from 4°C to 37°C and for up to 2 weeks at 50°C.


Asunto(s)
Estabilidad de Medicamentos , Vacunas contra el Virus del Ébola/inmunología , Vacunas contra el Virus del Ébola/efectos de la radiación , Fiebre Hemorrágica Ebola/prevención & control , Vacunas Antirrábicas/inmunología , Vacunas Antirrábicas/efectos de la radiación , Rabia/prevención & control , Animales , Vacunas contra el Virus del Ébola/administración & dosificación , Vacunas contra el Virus del Ébola/genética , Femenino , Calor , Mesocricetus , Vacunas Antirrábicas/administración & dosificación , Vacunas Antirrábicas/genética , Temperatura , Resultado del Tratamiento , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/genética , Vacunas de Productos Inactivados/inmunología , Vacunas de Productos Inactivados/efectos de la radiación , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/efectos de la radiación , Volatilización
10.
J Gen Virol ; 100(8): 1222-1233, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31259681

RESUMEN

Rabies, caused by rabies virus (RABV), is a fatal zoonosis, which still poses a threat to public health in most parts of the world. Glycoprotein of RABV is the only viral surface protein, which is critical for the induction of virus-neutralizing antibodies (VNA). In order to improve the production of VNA, recombinant RABVs containing two copies of G gene and codon-optimized G gene were constructed by using reverse genetics, named LBNSE-dG and LBNSE-dOG, respectively. After being inoculated into the mouse brains, LBNSE-dOG induced more apoptosis and recruited more inflammatory cells than LBNSE-dG and LBNSE, resulting in reduced virulence in vivo. After intramuscular (im) immunization in mice, LBNSE-dOG promoted the formation of germinal centres (GCs), the recruitment of GC B cells and the generation of antibody-secreting cells (ASCs) in the draining lymph nodes (LNs). Consistently, LBNSE-dOG boosted the production of VNA and provided better protection against lethal RABV challenge than LBNSE-dG and LBNSE when it was used as both live and inactivated vaccines. Our results demonstrate that the codon-optimized RABV LBNSE-dOG displays attenuated pathogenicity and enhanced immunogenicity, therefore it could be a potential candidate for the next generation of rabies vaccines.


Asunto(s)
Codón/genética , Glicoproteínas/genética , Glicoproteínas/inmunología , Inmunidad Humoral , Virus de la Rabia/inmunología , Rabia/inmunología , Proteínas Virales/genética , Proteínas Virales/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Femenino , Glicoproteínas/administración & dosificación , Humanos , Ratones , Ratones Endogámicos ICR , Rabia/prevención & control , Rabia/virología , Vacunas Antirrábicas/administración & dosificación , Vacunas Antirrábicas/genética , Vacunas Antirrábicas/inmunología , Virus de la Rabia/genética , Proteínas Virales/administración & dosificación
11.
J Virol ; 91(3)2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27852854

RESUMEN

Rabies remains a public health threat in most parts of the world, and approximately 99% of the cases are transmitted by dogs. There is an urgent need to develop an efficacious and affordable vaccine to control canine-transmitted rabies in developing countries. Our previous studies demonstrate that overexpression of chemokines/cytokines such as CCL-3 (MIP-1α) and granulocyte-macrophage colony-stimulating factor (GM-CSF) can enhance the immunogenicity of rabies vaccines. In the present study, the chemokine CXCL13 was inserted into the genome of the recombinant rabies virus (rRABV) strain LBNSE, and the effect of the chemokine CXCL13 on the immunogenicity of RABV was investigated. It was found that LBNSE-CXCL13 recruited follicular helper T (Tfh) and germinal center (GC) B cells, promoted the formation of GCs, and increased the population of plasma cells in immunized mice. Further studies showed that mice immunized with LBNSE-CXCL13 produced more rabies virus-neutralizing antibodies (VNAs) and developed better protection than those immunized with the parent virus LBNSE or the GM-CSF-expressing RABV (LBNSE-GM-CSF). Collectively, these findings provide a better understanding of the role of CXCL13 expression in the immunogenicity of the RABV, which may help in designing more-efficacious rabies vaccines. IMPORTANCE: Rabies is endemic in most parts of the world, and more effort is needed to develop affordable and effective vaccines to control or eliminate this disease. The chemokine CXCL13 recruits both Tfh and B cells, which is essential for the homing of Tfh cells and the development of B cell follicles. In this study, the effect of the overexpression of CXCL13 on the immunogenicity of the RABV was evaluated in a mouse model. We found that CXCL13 expression promoted humoral immunity by recruiting Tfh and GC B cells, facilitating the formation of GCs, and increasing the number of plasma cells. As expected, the overexpression of CXCL13 resulted in enhanced virus-neutralizing antibody (VNA) production and protection against a virulent RABV challenge. These findings provide a better understanding of the role of CXCL13 in RABV-induced immune responses, which will help in designing more efficacious rabies vaccines.


Asunto(s)
Linfocitos B/inmunología , Quimiocina CXCL13/genética , Expresión Génica , Centro Germinal/inmunología , Inmunidad Humoral , Vacunas Antirrábicas/genética , Vacunas Antirrábicas/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Linfocitos B/metabolismo , Quimiocina CXCL13/sangre , Quimiotaxis/inmunología , Cricetinae , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Ratones , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Rabia/inmunología , Rabia/prevención & control , Virus de la Rabia/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo
12.
J Virol ; 91(8)2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28148792

RESUMEN

Vaccine-induced B cells differentiate along two pathways. The follicular pathway gives rise to germinal centers (GCs) that can take weeks to fully develop. The extrafollicular pathway gives rise to short-lived plasma cells (PCs) that can rapidly secrete protective antibodies within days of vaccination. Rabies virus (RABV) postexposure prophylaxis (PEP) requires rapid vaccine-induced humoral immunity for protection. Therefore, we hypothesized that targeting extrafollicular B cell responses for activation would improve the speed and magnitude of RABV PEP. To test this hypothesis, we constructed, recovered, and characterized a recombinant RABV-based vaccine expressing murine B cell activating factor (BAFF) (rRABV-mBAFF). BAFF is an ideal molecule to improve early pathways of B cell activation, as it links innate and adaptive immunity, promoting potent B cell responses. Indeed, rRABV-mBAFF induced a faster, higher antibody response in mice and enhanced survivorship in PEP settings compared to rRABV. Interestingly, rRABV-mBAFF and rRABV induced equivalent numbers of GC B cells, suggesting that rRABV-mBAFF augmented the extrafollicular B cell pathway. To confirm that rRABV-mBAFF modulated the extrafollicular pathway, we used a signaling lymphocytic activation molecule (SLAM)-associated protein (SAP)-deficient mouse model. In response to antigen, SAP-deficient mice form extrafollicular B cell responses but do not generate GCs. rRABV-mBAFF induced similar anti-RABV antibody responses in SAP-deficient and wild-type mice, demonstrating that BAFF modulated immunity through the extrafollicular and not the GC B cell pathway. Collectively, strategies that manipulate pathways of B cell activation may facilitate the development of a single-dose RABV vaccine that replaces current complicated and costly RABV PEP.IMPORTANCE Effective RABV PEP is currently resource- and cost-prohibitive in regions of the world where RABV is most prevalent. In order to diminish the requirements for rabies immunoglobulin (RIG) and multiple vaccinations for effective prevention of clinical rabies, a more rapidly protective vaccine is needed. This work presents a successful approach to rapidly generate antibody-secreting PCs in response to vaccination by targeting the extrafollicular B cell pathway. We demonstrate that the improved early antibody responses induced by rRABV-mBAFF confer improved protection against RABV in a PEP model. Significantly, activation of the early extrafollicular B cell pathway, such as that demonstrated here, could improve the efficacy of vaccines targeting other pathogens against which rapid protection would decrease morbidity and mortality.


Asunto(s)
Linfocitos B/fisiología , Diferenciación Celular , Profilaxis Posexposición/métodos , Vacunas Antirrábicas/inmunología , Rabia/prevención & control , Animales , Anticuerpos Antivirales/sangre , Factor Activador de Células B/genética , Factor Activador de Células B/metabolismo , Linfocitos B/inmunología , Ratones , Vacunas Antirrábicas/administración & dosificación , Vacunas Antirrábicas/genética , Análisis de Supervivencia , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
13.
Mol Biol (Mosk) ; 52(3): 527-532, 2018.
Artículo en Ruso | MEDLINE | ID: mdl-29989585

RESUMEN

The possibility of enhancing the immunogenicity of the rabies virus glycoprotein antigen encoded by a DNA vaccine has been investigated. Ubiquitin-like protein FAT10 has been attached to the N-terminus of the glycoprotein to target it to the proteasome and stimulate its presentation by MHC class I. Two forms of the protein, chimeric and original, have been detected in cells transfected with the DNA construct encoding the chimeric protein. The presence of the glycoprotein on the cell surface has been detected by immunostaining of transfected cells. The production of IgG and IgG2a antibodies has been more efficiently induced in mice immunized with the plasmid that encodes the chimeric protein than in those immunized with the plas-mid that encodes unmodified glycoprotein. Moreover, the level of IgG2a antibodies exceeded the level of IgG1 antibodies, which indicates a preferential increase in the Th1 component of the immune response. The proposed DNA construct that encodes a modified glycoprotein with a proteasome degradation signal maybe a promising DNA vaccine immunogen for post-exposure prophylaxis of rabies.


Asunto(s)
Anticuerpos Antivirales/inmunología , ADN Viral , Inmunoglobulina G/inmunología , Glicoproteínas de Membrana , Complejo de la Endopetidasa Proteasomal , Señales de Clasificación de Proteína/genética , Vacunas Antirrábicas , Virus de la Rabia , Proteínas Virales , Animales , Formación de Anticuerpos , ADN Viral/genética , ADN Viral/inmunología , Femenino , Células HeLa , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/inmunología , Vacunas Antirrábicas/genética , Vacunas Antirrábicas/inmunología , Virus de la Rabia/genética , Virus de la Rabia/inmunología , Proteínas Virales/genética , Proteínas Virales/inmunología
14.
Mol Biol (Mosk) ; 52(2): 314-317, 2018.
Artículo en Ruso | MEDLINE | ID: mdl-29695700

RESUMEN

Safe and effective anti-rabies vaccines are intensely sought worldwide. DNA vaccines have already shown their efficacy and safety and have occupied a special place in the field. Two prototype anti-rabies DNA vaccines were compared for the potential to induce virus-specific antibody production. One vector contained a codon-optimized gene with a territory-adapted consensus sequence of the rabies virus glycoprotein. The other one expressed the same glycoprotein in fusion with a c-CD63 lysosome targeting motif at the C terminus. ELISA of serum samples from immunized mice showed that the c-CD63 variant induced more efficient antibody production and shifted the IgG2a/IgG1 ratio towards the Th2-type immune response. The results gave grounds to believe that the approach successfully applied to the rabies glycoprotein may help to develop new-generation anti-rabies vaccines.


Asunto(s)
Anticuerpos Antivirales/inmunología , Formación de Anticuerpos/efectos de los fármacos , Inmunoglobulina G/inmunología , Señales de Clasificación de Proteína , Vacunas Antirrábicas , Vacunas de ADN , Proteínas Virales , Secuencia de Aminoácidos , Animales , Femenino , Glicoproteínas/genética , Glicoproteínas/inmunología , Ratones , Ratones Endogámicos BALB C , Transporte de Proteínas/genética , Transporte de Proteínas/inmunología , Vacunas Antirrábicas/genética , Vacunas Antirrábicas/inmunología , Vacunas Antirrábicas/farmacología , Virus de la Rabia/genética , Virus de la Rabia/inmunología , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Vacunas de ADN/farmacología , Proteínas Virales/genética , Proteínas Virales/inmunología
15.
Virol J ; 14(1): 122, 2017 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-28646885

RESUMEN

BACKGROUND: Rabies is an important viral zoonosis that causes acute encephalitis and death in mammals. To date, several recombinant vaccines have been developed based on G protein, which is considered to be the main antigen, and these vaccines are used for rabies control in many countries. Most recombinant viruses expressing RABV G protein retain the G gene from attenuated RABV. Not enough is currently known about the protective effect against RABV of a combination of recombinant adenoviruses expressing the G and N proteins of pathogenic street RABV. METHODS: We constructed a recombinant adenovirus (Ad-0910Gsped) expressing the signal peptide and ectodomain (sped) of G protein of the Korean street strain, and evaluated the immunological protection conferred by a single and combination of three kinds of recombinant adenoviruses (Ad-0910Gsped and Ad-0910G with or without Ad-0910 N) in mice. RESULTS: A combination of Ad-0910G and Ad-0910 N conferred improved immunity against intracranial challenge compared to single administration of Ad-0910G. The Ad-0910G virus, expressing the complete G protein, was more immunogenic than Ad-0910Gsped, which expressed a truncated G protein with the transmembrane and cytoplasmic domains removed. Additionally, oral vaccination using a combination of viruses led to complete protection. CONCLUSIONS: Our results suggest that this combination of viruses is a viable new intramuscular and oral vaccine candidate.


Asunto(s)
Adenoviridae/genética , Antígenos Virales/inmunología , Portadores de Fármacos , Glicoproteínas/inmunología , Vacunas Antirrábicas/inmunología , Virus de la Rabia/inmunología , Rabia/prevención & control , Proteínas del Envoltorio Viral/inmunología , Administración Oral , Animales , Antígenos Virales/genética , Glicoproteínas/genética , Inyecciones Intramusculares , Ratones , Rabia/inmunología , Vacunas Antirrábicas/genética , Virus de la Rabia/genética , Resultado del Tratamiento , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/genética
16.
Arch Virol ; 162(2): 323-332, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27796547

RESUMEN

The rabies virus envelope glycoprotein (RVGP) is the main antigen of rabies virus and is the only viral component present in all new rabies vaccines being proposed. Many approaches have been taken since DNA recombinant technology became available to express an immunogenic recombinant rabies virus glycoprotein (rRVGP). These attempts are reviewed here, and the relevant results are discussed with respect to the general characteristics of the rRVGP, the expression system used, the expression levels achieved, the similarity of the rRVGP to the native glycoprotein, and the immunogenicity of the vaccine preparation. The most recent studies of rabies vaccine development have concentrated on in vivo expression of rRVGP by viral vector transduction, serving as the biotechnological basis for a new generation of rabies vaccines.


Asunto(s)
Anticuerpos Antivirales/biosíntesis , Antígenos Virales/inmunología , Inmunogenicidad Vacunal , Vacunas Antirrábicas/genética , Virus de la Rabia/inmunología , Proteínas del Envoltorio Viral/genética , Animales , Antígenos Virales/química , Antígenos Virales/genética , Línea Celular , Drosophila melanogaster/citología , Drosophila melanogaster/virología , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Vacunas Antirrábicas/administración & dosificación , Vacunas Antirrábicas/biosíntesis , Virus de la Rabia/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Spodoptera/citología , Spodoptera/virología , Vacunas Sintéticas , Proteínas del Envoltorio Viral/biosíntesis , Proteínas del Envoltorio Viral/inmunología
17.
Mol Biol (Mosk) ; 51(3): 460-463, 2017.
Artículo en Ruso | MEDLINE | ID: mdl-28707662

RESUMEN

The glycoprotein of rabies virus is the central antigen elicited the immune response to infection; therefore, the majority of developing anti-rabies vaccines are based on this protein. In order to increase the efficacy of DNA immunogen encoding rabies virus glycoprotein, the construction of chimeric protein with the CD63 domain has been proposed. The CD63 is a transmembrane protein localized on the cell surface and in lysosomes. The lysosome targeting motif GYEVM is located at its C-terminus. We used the domain that bears this motif (c-CD63) to generate chimeric glycoprotein in order to relocalize it into lysosomes. Here, it was shown that, in cells transfected with plasmid that encodes glycoprotein with c-CD63 motif at the C-terminus, the chimeric protein was predominantly observed in lysosomes and at the cell membrane where the unmodified glycoprotein is localized in the endoplasmic reticulum and at the cell surface. We suppose that current modification of the glycoprotein may improve the immunogenicity of anti-rabies DNA vaccines due to more efficient antibody production.


Asunto(s)
Glicoproteínas/genética , Vacunas Antirrábicas/genética , Rabia/inmunología , Tetraspanina 30/genética , Glicoproteínas/inmunología , Células HeLa , Humanos , Lisosomas/genética , Lisosomas/inmunología , Dominios Proteicos/genética , Dominios Proteicos/inmunología , Rabia/prevención & control , Rabia/virología , Vacunas Antirrábicas/inmunología , Virus de la Rabia/inmunología , Virus de la Rabia/patogenicidad , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Tetraspanina 30/inmunología , Vacunas de ADN/inmunología , Vacunas de ADN/uso terapéutico
18.
J Gen Virol ; 97(12): 3154-3160, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27902338

RESUMEN

Previous studies have demonstrated that the lack of interleukin-21 (IL-21) signalling could affect specific antibody induction after rabies vaccination. Here, to further investigate the over-expression of IL-21 on the immunogenicity of rabies virus (RABV), a recombinant RABV expressing murine IL-21, designated LBNSE-IL21, was constructed and evaluated in a mouse model. It was found that in mice immunized with LBNSE-IL21, there was a substantial increase in the number of T follicular helper cells and germinal centre B cells but no enhancement of dendritic cell activation. Furthermore, significantly higher rabies virus-neutralizing antibody (VNA) titres were produced in mice immunized with LBNSE-IL21 than in mice immunized with the parent virus LBNSE in the first six weeks, resulting in higher protection. Together, these results suggest that LBNSE-IL21 can induce a rapid and robust VNA titre, and it has the potential to be developed as a promising rabies vaccine.


Asunto(s)
Centro Germinal/inmunología , Interleucinas/inmunología , Vacunas Antirrábicas/inmunología , Virus de la Rabia/inmunología , Rabia/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Anticuerpos Antivirales/inmunología , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Interleucinas/administración & dosificación , Interleucinas/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Rabia/prevención & control , Rabia/virología , Vacunas Antirrábicas/administración & dosificación , Vacunas Antirrábicas/genética , Virus de la Rabia/genética
19.
J Virol ; 89(6): 3427-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25552723

RESUMEN

Rabies remains a major public health threat around the world. Once symptoms appear, there is no effective treatment to prevent death. In this work, we tested a recombinant parainfluenza virus 5 (PIV5) strain expressing the glycoprotein (G) of rabies (PIV5-G) as a therapy for rabies virus infection: we have found that PIV5-G protected mice as late as 6 days after rabies virus infection. PIV5-G is a promising vaccine for prevention and treatment of rabies virus infection.


Asunto(s)
Vectores Genéticos/genética , Virus de la Parainfluenza 5/genética , Vacunas Antirrábicas/administración & dosificación , Virus de la Rabia/inmunología , Rabia/prevención & control , Proteínas del Envoltorio Viral/administración & dosificación , Animales , Anticuerpos Antivirales/inmunología , Expresión Génica , Vectores Genéticos/metabolismo , Humanos , Ratones , Virus de la Parainfluenza 5/metabolismo , Rabia/inmunología , Rabia/virología , Vacunas Antirrábicas/genética , Vacunas Antirrábicas/inmunología , Virus de la Rabia/genética , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
20.
Arch Virol ; 161(10): 2863-70, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27438075

RESUMEN

Two recombinant rabies viruses overexpressing their glycoprotein (G) were compared in this study, with the overexpressed G inserted between P and M genes (named LBNSE-PM-G), and between the G and L genes (named LBNSE-GL-G), respectively. LBNSE-PM-G produced more G protein and induced stronger apoptosis than LBNSE-GL-G in infected cells, while the amount of virion-incorporated G in LBNSE-PM-G was less than in LBNSE-GL-G. Mice immunized with inactivated LBNSE-PM-G produced lower titers of virus-neutralizing antibody, and this recombinant conferred worse protection than LBNSE-GL-G. Our results suggest that over expressed G gene inserted between G and L, but not between P and M, enhanced the immunogenicity when used as an inactivated rabies vaccine.


Asunto(s)
Antígenos Virales/inmunología , Glicoproteínas/inmunología , Vacunas Antirrábicas/inmunología , Virus de la Rabia/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Antígenos Virales/genética , Modelos Animales de Enfermedad , Femenino , Glicoproteínas/genética , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Rabia/prevención & control , Vacunas Antirrábicas/genética , Virus de la Rabia/genética , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda