Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Microbes Infect ; 26(4): 105321, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38461968

RESUMO

Rabies virus (RABV) is a lethal neurotropic virus that causes 60,000 human deaths every year globally. RABV infection is characterized by the suppression of the interferon (IFN)-mediated antiviral response. However, molecular mechanisms leading to RABV sensing by RIG-I-like receptors (RLR) that initiates IFN signaling currently remain elusive. Here, we showed that RABV RNAs are primarily recognized by the RIG-I RLR, resulting in an IFN response in the infected cells, but this response varied according to the type of RABV used. Pathogenic RABV strain RNAs, Tha, were poorly detected in the cytosol by RIG-I and therefore caused a weak antiviral response. However, we revealed a strong IFN activity triggered by the attenuated RABV vaccine strain RNAs, SAD, mediated by RIG-I. We characterized two major 5' copy-back defective interfering (5'cb DI) genomes generated during SAD replication. Furthermore, we identified an interaction between 5'cb DI genomes, and RIG-I correlated with a high stimulation of the type I IFN signaling. This study indicates that wild-type RABV RNAs poorly activate the RIG-I pathway, while the presence of 5'cb DIs in the live-attenuated vaccine strain serves as an intrinsic adjuvant that strengthens its efficiency by enhancing RIG-I detection thus strongly stimulates the IFN response.


Assuntos
Proteína DEAD-box 58 , Vírus da Raiva , Humanos , Linhagem Celular , Proteína DEAD-box 58/metabolismo , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/imunologia , Interferon Tipo I/metabolismo , Interferon Tipo I/imunologia , Raiva/imunologia , Raiva/virologia , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Vírus da Raiva/genética , Vírus da Raiva/patogenicidade , Receptores Imunológicos/metabolismo , RNA Viral/genética , Transdução de Sinais , Replicação Viral
2.
J Virol ; 95(23): e0141421, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34495701

RESUMO

Rabies, caused by rabies virus (RABV), remains a serious threat to public health in most countries worldwide. At present, the administration of rabies vaccines has been the most effective strategy to control rabies. Herein, we evaluate the effect of colloidal manganese salt (Mn jelly [MnJ]) as an adjuvant of rabies vaccine in mice, cats, and dogs. The results showed that MnJ promoted type I interferon (IFN-I) and cytokine production in vitro and the maturation of dendritic cells (DCs) in vitro and in vivo. Besides, MnJ serving as an adjuvant for rabies vaccines could significantly facilitate the generation of T follicular helper (Tfh) cells, germinal center (GC) B cells, plasma cells (PCs), and RABV-specific antibody-secreting cells (ASCs), consequently improve the immunogenicity of rabies vaccines, and provide better protection against virulent RABV challenge. Similarly, MnJ enhanced the humoral immune response in cats and dogs as well. Collectively, our results suggest that MnJ can facilitate the maturation of DCs during rabies vaccination, which can be a promising adjuvant candidate for rabies vaccines. IMPORTANCE Extending the humoral immune response by using adjuvants is an important strategy for vaccine development. In this study, a novel adjuvant, MnJ, supplemented in rabies vaccines was evaluated in mice, cats, and dogs. Our results in the mouse model revealed that MnJ increased the numbers of mature DCs, Tfh cells, GC B cells, PCs, and RABV-specific ASCs, resulting in enhanced immunogenicity and protection rate of rabies vaccines. We further found that MnJ had the same stimulative effect in cats and dogs. Our study provides the first evidence that MnJ serving as a novel adjuvant of rabies vaccines can boost the immune response in both a mouse and pet model.


Assuntos
Adjuvantes Imunológicos , Manganês/farmacologia , Vacina Antirrábica/imunologia , Animais , Anticorpos Antivirais/sangue , Células Produtoras de Anticorpos/imunologia , Linfócitos B/imunologia , Linfócitos T CD4-Positivos , Gatos , Células Dendríticas/imunologia , Modelos Animais de Doenças , Cães , Feminino , Centro Germinativo/imunologia , Imunidade Humoral , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Plasmócitos/imunologia , Raiva/imunologia , Vírus da Raiva/imunologia , Vacinação , Desenvolvimento de Vacinas
3.
J Virol ; 94(13)2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32321812

RESUMO

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.


Assuntos
Anticorpos Neutralizantes/uso terapêutico , Avaliação Pré-Clínica de Medicamentos/métodos , Vírus da Raiva/imunologia , Animais , Anticorpos Antivirais/imunologia , Antivirais/farmacologia , Linhagem Celular , Proteção Cruzada , Humanos , Biblioteca de Peptídeos , Raiva/prevenção & controle , Vacina Antirrábica/imunologia , Vírus da Raiva/metabolismo , Vírus da Raiva/patogenicidade , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/imunologia , Vesiculovirus/genética , Vesiculovirus/imunologia , Proteínas Virais de Fusão/farmacologia
4.
Vector Borne Zoonotic Dis ; 20(4): 303-309, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31794689

RESUMO

Vaccination against rabies and routine antibody testing of subjects participating in programs for the surveillance and control of rabies in animals is strongly recommended. The scope of this study is to describe the antibody level as measured by a commercial enzyme-linked immunosorbent assay (ELISA) after primary and booster intramuscular vaccination with a purified vero-cell rabies vaccine (PVRV) in high-risk professionals and to determine the influence of an array of factors on antibody level, that is, time elapsed since primary immunization series and booster dose, sex, age, pathologic conditions, high-risk occupation, and peak antibody level after initial scheme and booster dose. A primary series of three doses of PVRV was administered and a commercial ELISA was recommended 14 days postimmunization with continuous repetition at 6 months and yearly intervals for the laboratory personnel and the rest of the professionals, respectively. The protective antibody titer was defined as a minimum of 0.5 equivalent units/mL (EU/mL) (seroconvertion) and a booster dose was applied if the titer was determined nonprotective. The seroconversion rate (SCR) after primary vaccination was 100%, with a geometric mean titer (GMT) of 2.90 EU/mL (interquartile range [IQR]: 1.85-3.45). After booster vaccination due to nonprotective titer, the SCR was 100% and the GMT increased by 678% (95% confidence interval [CI]: 514-887) reaching 4.25 EU/mL (IQR: 4.00-4.60), 2.5 times higher than the GMT elicited by the primary vaccine scheme in the respective recipients. The titer dropped by 1.20% per month (95% CI: 0.52-1.89) regardless of booster administration or any other factor. Women had 51% higher titer compared with men (95% CI: 6-116). High-risk professionals should be verified for adequate antibody titers, but routine administration of a single booster dose of PVRV 1 year after the primary series could be considered; more evidence is needed to support the benefit in terms of immunity and logistics.


Assuntos
Anticorpos Antivirais/sangue , Exposição Ocupacional/prevenção & controle , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Raiva/prevenção & controle , Animais , Animais Selvagens , Raposas/virologia , Humanos , Programas Nacionais de Saúde , Vigilância da População , Profilaxia Pré-Exposição , Vacina Antirrábica/administração & dosagem , Fatores de Risco , Vacinação , Médicos Veterinários
5.
Virol Sin ; 34(6): 662-672, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31254272

RESUMO

Rabies is a zoonotic disease that still causes 59,000 human deaths each year, and rabies vaccine is the most effective way to control the disease. Our previous studies suggested that the maturation of DC plays an important role in enhancing the immunogenicity of rabies vaccine. Flt3L has been reported to own the ability to accelerate the DC maturation, therefore, in this study, a recombinant rabies virus expressing mouse Flt3L, designated as LBNSE-Flt3L, was constructed, and its immunogenicity was characterized. It was found that LBNSE-Flt3L could enhance the maturation of DC both in vitro and in vivo, and significantly more TFH cells and Germinal Center B (GC B) cells were generated in mice immunized with LBNSE-Flt3L than those immunized with the parent virus LBNSE. Consequently, expressing of Flt3L could elevate the level of virus-neutralizing antibodies (VNA) in immunized mice which provides a better protection from a lethal rabies virus challenge. Taken together, our study extends the potential of Flt3L as a good adjuvant to develop novel rabies vaccine by enhancing the VNA production through activating the DC-TFH-GC B axis in immunized mice.


Assuntos
Adjuvantes Imunológicos , Proteínas de Membrana/imunologia , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Raiva/imunologia , Adjuvantes Imunológicos/genética , Adjuvantes Imunológicos/metabolismo , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Linfócitos B/imunologia , Células Dendríticas/imunologia , Feminino , Centro Germinativo/imunologia , Imunização , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Plasmócitos/imunologia , Raiva/prevenção & controle , Vacina Antirrábica/genética , Taxa de Sobrevida , Linfócitos T Auxiliares-Indutores/imunologia , Vacinas Sintéticas
6.
Vaccine ; 35(49 Pt B): 6898-6904, 2017 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-28899628

RESUMO

The RepliVax® vaccine (RV) platform is based on flavivirus genomes that are rationally attenuated by deletion. These single-cycle RV vaccine candidates targeting flavivirus pathogens have been demonstrated to be safe, highly immunogenic, and efficacious in animal models, including non-human primates. Here we show utility of the technology for delivery of a non-flavivirus immunogen by engineering several West Nile-based RV vectors to express full-length rabies virus G protein. The rabies virus G protein gene was incorporated in place of different West Nile structural protein gene deletions. The resulting RV-RabG constructs were demonstrated to replicate to high titers (8 log10 infectious particles/ml) in complementing helper cells. Following infection of normal cells, they provided efficient rabies virus G protein expression, but did not spread to surrounding cells. Expression of rabies virus G protein was stable and maintained through multiple rounds of in vitro passaging. A sensitive neurovirulence test in 2-3 day old neonatal mice demonstrated that RV-RabG candidates were completely avirulent indicative of high safety. We evaluated the RV-RabG variants in several animal models (mice, dogs, and pigs) and demonstrated that a single dose elicited high titers of rabies virus-neutralizing antibodies and protected animals from live rabies virus challenge (mice and dogs). Importantly, dogs were protected at both one and two years post-immunization, demonstrating durable protective immunity. The data demonstrates the potential of the RepliVax® technology as a potent vector delivery platform for developing vaccine candidates against non-flavivirus targets.


Assuntos
Flavivirus/genética , Vetores Genéticos , Vacina Antirrábica/genética , Vacinas Sintéticas/imunologia , Proteínas do Envelope Viral , Vacinas Virais/imunologia , Animais , Animais Recém-Nascidos , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Modelos Animais de Doenças , Cães , Avaliação Pré-Clínica de Medicamentos , Feminino , Camundongos , Raiva/prevenção & controle , Vacina Antirrábica/administração & dosagem , Vacina Antirrábica/química , Vacina Antirrábica/imunologia , Vírus da Raiva/química , Vírus da Raiva/imunologia , Suínos , Vacinação , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem
7.
Virology ; 489: 165-72, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26765968

RESUMO

Vaccination alone is not sufficiently effective to protect human from post-exposure rabies virus infection due to delayed generation of rabies virus neutralizing antibodies and weak cellular immunity. Therefore, it is vital to develop safer and more efficacious vaccine against rabies. PIKA, a stabilized chemical analog of double-stranded RNA that interacts with TLR3, was employed as adjuvant of rabies vaccine. The efficacy and safety of PIKA rabies vaccine were evaluated. The results showed that PIKA rabies vaccine enhanced both humoral and cellular immunity. After viral challenge, PIKA rabies vaccine protected 70-80% of animals, while the survival rate of non-adjuvant vaccine group (control) was 20-30%. According to the results of toxicity tests, PIKA and PIKA rabies vaccine are shown to be well tolerated in mice. Thus, this study indicates that PIKA rabies vaccine is an effective and safe vaccine which has the potential to develop next-generation rabies vaccine and encourage the start of clinical studies.


Assuntos
RNA de Cadeia Dupla/imunologia , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Raiva/imunologia , Receptor 3 Toll-Like/agonistas , Animais , Anticorpos Antivirais/imunologia , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Imunidade Celular , Masculino , Camundongos , Camundongos Endogâmicos BALB C , RNA de Cadeia Dupla/administração & dosagem , RNA de Cadeia Dupla/efeitos adversos , RNA de Cadeia Dupla/genética , Raiva/prevenção & controle , Raiva/virologia , Vacina Antirrábica/administração & dosagem , Vacina Antirrábica/efeitos adversos , Vírus da Raiva/genética , Receptor 3 Toll-Like/imunologia
8.
J Infect Dis ; 212 Suppl 2: S414-24, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26063224

RESUMO

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.


Assuntos
Filoviridae/imunologia , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Raiva/imunologia , Vacinas de Produtos Inativados/imunologia , Animais , Anticorpos Antivirais/imunologia , Formação de Anticorpos/imunologia , Chlorocebus aethiops , Avaliação Pré-Clínica de Medicamentos/métodos , Vacinas contra Ebola/imunologia , Ebolavirus/imunologia , Glicoproteínas/imunologia , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/virologia , Macaca fascicularis , Marburgvirus/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Raiva/virologia , Sudão , Vacinação/métodos , Células Vero
9.
Int Immunopharmacol ; 20(2): 283-9, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24680943

RESUMO

The inactivated rabies virus vaccine (RV) is a relatively expensive vaccine, prone to failure in some cases. Ginsenoside Re (Re) is a saponin isolated from Panax ginseng, and has an adjuvant property. Here the adjuvant effect of Re to improve the immune response to the RV is evaluated in mice. ICR mice were immunized with saline, 2.50mg/kg Re, 20µl RV, 100µl RV, or 20µl of RV adjuvanted with Re (1.25, 2.50 or 5.00mg/kg). Different time points after boosting, we measured serum antibodies in blood samples and separated splenocytes to detect lymphocyte proliferation and the production of IL-4, IL-10, IL-12, and IFN-γ in vitro. We also compared immunizations containing 20µl RV and 20µl RV adjuvanted with Re (5.00mg/kg) for the expression of CD4(+) and CD8(+) T-cell subsets at different time points. Results indicated that co-administration of Re significantly enhanced serum antibody titers, increased the CD4(+):CD8(+) ratio, and enhanced both proliferation responses and IL-4, IL-10, IL-12 and IFN-γ secretions. Both Th1 and Th2 immune responses were activated. The supplementation of the Re (5.00mg/kg) to 20µl of RV significantly amplified serum antibody responses and Th1/Th2 responses inducing similar protection as did 100µl of RV. This suggests that Re could be used to reduce the dose, and therefore the cost, of the RV to achieve the same effective protection. Re merits further studies for use with vaccines of mixed Th1/Th2 immune responses.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Ginsenosídeos/administração & dosagem , Panax , Vacina Antirrábica , Vírus da Raiva/imunologia , Raiva/imunologia , Células Th1/efeitos dos fármacos , Células Th2/efeitos dos fármacos , Animais , Anticorpos Antivirais/sangue , Formação de Anticorpos/efeitos dos fármacos , Relação CD4-CD8 , Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Feminino , Imunização , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , Células Th1/imunologia , Células Th2/imunologia , Vacinas de Produtos Inativados
10.
Microbes Infect ; 16(3): 196-202, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24309427

RESUMO

Extracellular nucleotides such as adenosine 5'-triphospate (ATP) and uridine 5'-triphosphate (UTP) interact with P2 purinergic receptors on the surface of phagocytic cells and induce various physiological reactions. In this study, the production of antibody in mice immunized with an inactivated rabies vaccine containing these nucleotides was investigated. Injection of inactivated rabies vaccine with UTP, but not with ATP, induced significantly higher serum antibody production in mice. The enhancement of antibody production by UTP was inhibited by an anti-P2Y4 receptor antibody. In an air pouch experiment, UTP treatment increased the number of monocytes and macrophages infiltrating the pouch and up-regulated the gene expression of IL-4 and IL-13 in the regional lymph nodes. These results suggested that UTP admixed with rabies vaccine activates Th2 cells and induces a humoral immune response. Furthermore, the survival rate of mice immunized with a rabies vaccine admixed with UTP before rabies virus challenge was slightly higher than that of control mice. In conclusion, UTP can act as a vaccine adjuvant to enhance antibody production against the rabies virus in mice.


Assuntos
Adjuvantes Imunológicos/farmacologia , Anticorpos Antivirais/sangue , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Uridina Trifosfato/imunologia , Uridina Trifosfato/farmacologia , Trifosfato de Adenosina , Adjuvantes Imunológicos/química , Animais , Formação de Anticorpos/efeitos dos fármacos , Formação de Anticorpos/imunologia , Feminino , Interleucinas/análise , Interleucinas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Vacina Antirrábica/administração & dosagem , Vacina Antirrábica/química , Vacina Antirrábica/farmacologia , Uridina Trifosfato/química
11.
Vaccine ; 26(52): 6936-44, 2008 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-18940223

RESUMO

We tested two post-exposure prophylaxes (PEPs) for rabies in laboratory animals; one was a traditional antirabies vaccine for humans via intramuscular route (IM), and the other was a DNA vaccine administered by intranasal route (IN). In contrast to The World Health Organization's recommended five-dose PEP, we gave only four doses without hyper-immune antirabies sera, making the PEP more rigorous. All animals were challenged with challenge virus strain (CVS); 16h later, PEP was applied. All animals that received the PEP with DNA/IN survived, and 87% of the rabbits and 80% of the mice that received the PEP with traditional antirabies vaccine/IM survived. Negative controls succumbed to infection. The expression of G protein was detected in the NALT, cerebellum, cerebral cortex (neocortex), cerebellum and hippocampus, mainly in the glial cells (microglia) and microvessels. On the other hand, plasmid construct was detected in brain and its mRNA expression in medium and posterior encephalon. The efficiency of this DNA/IN PEP is probably due to the early expression of the antigen in the brain stimulating the immune system locally.


Assuntos
Vacina Antirrábica/uso terapêutico , Raiva/prevenção & controle , Administração Intranasal , Animais , Encéfalo/imunologia , Encéfalo/patologia , Encéfalo/virologia , Linhagem Celular , Corantes , Cricetinae , DNA Complementar/biossíntese , DNA Complementar/genética , Amarelo de Eosina-(YS) , Feminino , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/imunologia , Hematoxilina , Esquemas de Imunização , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Plasmídeos/genética , RNA Viral/genética , RNA Viral/imunologia , Coelhos , Vacina Antirrábica/administração & dosagem , Vacina Antirrábica/imunologia , Vírus da Raiva/genética , Vírus da Raiva/imunologia , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia , Vacinas de DNA/uso terapêutico
14.
Microbes Infect ; 8(4): 1090-7, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16524754

RESUMO

Safe and effective vaccination is important for rabies prevention in animals. Although several genetically engineered rabies vaccines have been developed, few have been licensed for use, principally due to biosafety concerns or due to poor efficacy in animal models. In this paper, we describe the construction and characterization of a replication-competent recombinant canine adenovirus type-2 expressing the rabies virus glycoprotein (SRV9 strain) by a different strategy from that reported previously, i.e., the recombinant genome carrying the glycoprotein cDNA was generated by a series of strictly gene cloning steps, infectious recombinant virus was obtained by transfecting the recombinant genome into a canine kidney cell line, MDCK. This recombinant virus, CAV-E3delta-CGS, was subcutaneously injected into dogs. All vaccinated dogs produced effective neutralizing antibodies after one inoculation and a stronger anamnestic immune response was produced after booster injection. The immunized dogs could survive the challenge of 60,000 mouse LD50 CVS-24, which is lethal to all unimmunized dogs and is comparable to the conventional vaccines. The immunity lasts for months with a protective level of neutralizing antibody. This recombinant virus would be an alternative to the attenuated and the inactivated rabies vaccines and be prospective in immunizing dogs against rabies.


Assuntos
Adenovirus Caninos/genética , Vetores Genéticos , Glicoproteínas/genética , Vacina Antirrábica/administração & dosagem , Raiva/prevenção & controle , Vírus Reordenados/genética , Proteínas Virais/genética , Animais , Anticorpos Antivirais/sangue , Cães , Avaliação Pré-Clínica de Medicamentos , Esquemas de Imunização , Injeções Subcutâneas , Camundongos , Testes de Neutralização , Raiva/imunologia , Vírus da Raiva/imunologia , Vacinas de DNA/administração & dosagem
15.
J Nutr ; 132(6 Suppl 2): 1720S-4S, 2002 06.
Artigo em Inglês | MEDLINE | ID: mdl-12042506

RESUMO

We studied the effects of feeding an antioxidant blend of vitamins, minerals and carotenoids to a mixed adult dog population (n = 40, mean 4.4 +/- 1.85 y) for a 16-wk period. Compared to the control group of dogs (n = 20), the antioxidant (AOX)-supplemented group of dogs (n = 20) demonstrated significant increases in plasma levels of vitamin E and taurine by 4 wk of supplementation (P < 0.01) and total antioxidant activity (as measured by ferric-reducing antioxidant power assay) by 8 wk of supplementation (P < 0.05). Following 8 wk of supplementation, the AOX-supplemented dogs also showed significant reductions in both endogenous and exogenous DNA damage (P < 0.005) compared to that of the control dogs, as measured by the comet assay. Over an 8-wk rabies vaccination course that started at 8 wk supplementation, the AOX-supplemented dogs also demonstrated significantly higher vaccine-specific virus-neutralizing antibody levels at 2, 4 and 6 wk postvaccination (P < 0.05) and a tendency toward establishing a vaccine-specific antibody response quicker than did the control group of dogs. These findings in dogs suggest that antioxidant supplementation can achieve sustained increases in circulating levels of antioxidants that exert a protective effect by a decrease in DNA damage, leading to improved immunological performance. These findings also have implications in a wider context where free-radical damage has been associated with a variety of degenerative disorders and the aging process in general.


Assuntos
Antioxidantes/administração & dosagem , Dano ao DNA/efeitos dos fármacos , Cães/fisiologia , Animais , Anticorpos Antivirais/imunologia , Formação de Anticorpos/efeitos dos fármacos , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Dieta , Cães/sangue , Raiva/prevenção & controle , Vírus da Raiva/imunologia , Fatores de Tempo , Vacinação
16.
J Virol Methods ; 101(1-2): 85-94, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11849687

RESUMO

To assist in making recommendations for sampling of brains for the fluorescent antibody test (FAT), a study was conducted to determine the regions of the brain where rabies antigen is found most reliably. Each identifiable part of 252 rabies-positive brains of various species was re-tested using routine FA tests. It was found that there was frequent variation in the quantity of antigen between regions of the brain. The thalamus, pons and medulla were the most reliable parts of the brain as they were positive in all specimens tested. The cerebellum, hippocampus and different parts of the cerebrum were negative in, respectively, 4.5, 4.9 and 3.9-11.1% of positive brains. It is recommended that specimens for rabies diagnosis must include the brain stem. The structure of choice would be the thalamus as it was positive in all specimens and had the most frequent prevalence (97.8%) of abundant antigen. These findings contradict many old studies that state that the hippocampus should be the structure of choice for rabies diagnosis. The current data demonstrate that the reason for the old recommendations is that the hippocampus has the highest frequency of large inclusion bodies, as the reliability of the histological tests used previously depended on inclusion body size.


Assuntos
Antígenos Virais/análise , Encéfalo/virologia , Imunofluorescência/métodos , Vírus da Raiva/isolamento & purificação , Raiva/diagnóstico , Animais , Encéfalo/patologia , Carnívoros , Bovinos , Cerebelo/virologia , Equidae , Hipocampo/virologia , Bulbo/virologia , Ponte/virologia , Raiva/veterinária , Vírus da Raiva/imunologia , Reprodutibilidade dos Testes , Telencéfalo/virologia , Tálamo/virologia , Distribuição Tecidual
17.
East Mediterr Health J ; 7(4-5): 799-804, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-15332782

RESUMO

We tested the effects of vitamin A supplementation on the antibody titre of 40 healthy volunteers (age range: 10-35 years), who had received a complete course of antirabies vaccine (5 injections over 30 days). After determining the baseline serum vitamin A status of 80 volunteers, 20 pairs were matched for serum vitamin A level, body mass index, age, sex and socioeconomic status. One person from each pair was randomly assigned to an experimental or control group. The experimental group received vitamin A and antirabies vaccine. Controls received only the vaccine. The experimental group had significantly greater (2.1 times) serum antirabies titre than controls. This finding is an important step towards improving the economy of dosage of antirabies vaccines.


Assuntos
Formação de Anticorpos/efeitos dos fármacos , Vacina Antirrábica/imunologia , Vitamina A/uso terapêutico , Adolescente , Adulto , Anticorpos Antivirais/sangue , Anticorpos Antivirais/efeitos dos fármacos , Formação de Anticorpos/imunologia , Índice de Massa Corporal , Criança , Cromatografia Líquida de Alta Pressão , Terapia Combinada , Suplementos Nutricionais , Sinergismo Farmacológico , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imunocompetência/efeitos dos fármacos , Imunocompetência/imunologia , Masculino , Paquistão , Vacina Antirrábica/administração & dosagem , Vírus da Raiva/imunologia , Fatores Socioeconômicos , Inquéritos e Questionários , Resultado do Tratamento , Vacinação/métodos , Vitamina A/sangue , Vitamina A/imunologia , Deficiência de Vitamina A/sangue , Deficiência de Vitamina A/tratamento farmacológico , Deficiência de Vitamina A/imunologia
18.
Vaccine ; 17(3): 205-18, 1999 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-9987156

RESUMO

We investigated two forms (designated as yGI and yGII) of rabies virus glycoprotein (G) analogues produced in the G cDNA-transfected yeast cells. Molecular weights of yGI and yGII were estimated as 66 and 56 kDa, respectively, according to their relative mobility in SDS-PAGE. Although being produced in large amounts, yGI was present mostly in insoluble forms and hardly extractable with non-ionic detergents. The yGI reacted with polyclonal anti-G antibodies, but did not react with our conformational epitope-specific anti-G monoclonal antibodies (G-MAbs). No protective immunity was induced by yGI in guinea pigs nor in mice. On the other hand, yGII was Triton-soluble, but was only small in amount (at most 1% of total G proteins) and was shown to lack the cytoplasmic domain. The yGII, however, reacted with the G-MAbs and induced protective immunity in guinea pigs as well. When the G-cDNA was expressed in animal cells in culture, a single form (about 66 kDa) of G protein was produced, which displayed similar behaviors as seen in its reactivity with the MAbs and intracellular distribution as seen in the virus-infected cells. These results suggest that most G protein molecules were not processed normally in yeast cells, resulting in abnormal folding and multimer formation, while only a small fraction were occasionally folded normally to have conformational epitopes but were mostly deprived of the C-terminal portion.


Assuntos
Antígenos Virais/biossíntese , Glicoproteínas/imunologia , Vírus da Raiva/imunologia , Saccharomyces cerevisiae/metabolismo , Animais , Detergentes , Avaliação Pré-Clínica de Medicamentos , Regulação Viral da Expressão Gênica/fisiologia , Genes Virais , Vetores Genéticos , Glicosilação , Cobaias , Immunoblotting , Camundongos , Octoxinol , Solubilidade , Relação Estrutura-Atividade
19.
J Virol Methods ; 69(1-2): 63-72, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9504752

RESUMO

A rapid detection method for the six established genotypes of rabies and rabies-related viral RNA using RT-PCR-ELISA is described. The detection of digoxigenin-labelled amplified products is performed by solution hybridization to two specific, biotin-labelled, capture probes, which are complementary to the inner region of the amplification products. The capture probe and amplified product hybrid are then immobilised on a streptavidin-coated microtitre plate, bound products are detected by an anti-DIG Fab fragment conjugated to peroxidase, and colorimetric reaction automatically measured. This method was up to 100-fold more sensitive than Southern blot hybridization, detecting 0.00002 TCID50/ml of a genotype 1, classical rabies virus strain. The complete detection methodology from RT-PCR to PCR-ELISA detection could be completed within 10 h. Using this procedure, we were 100% successful in detecting 60 isolates from a representative selection of the six established genotypes from all over the world. This test is a useful additional tool for the detection of the rabies and rabies-related viruses, which is easy to perform, rapid and highly sensitive.


Assuntos
Ensaio de Imunoadsorção Enzimática , Lyssavirus/isolamento & purificação , Reação em Cadeia da Polimerase , Vírus da Raiva/isolamento & purificação , Raiva/diagnóstico , Animais , Southern Blotting , Genótipo , Humanos , Hibridização Genética , Lyssavirus/classificação , Lyssavirus/genética , Lyssavirus/imunologia , Raiva/veterinária , Vírus da Raiva/classificação , Vírus da Raiva/genética , Vírus da Raiva/imunologia , Reprodutibilidade dos Testes , Infecções por Rhabdoviridae/diagnóstico , Infecções por Rhabdoviridae/veterinária , Sensibilidade e Especificidade , Especificidade da Espécie
20.
J Cell Biol ; 126(2): 391-401, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7518464

RESUMO

The ERM family members, ezrin, radixin, and moesin, localizing just beneath the plasma membranes, are thought to be involved in the actin filament/plasma membrane association. To identify the integral membrane protein directly associated with ERM family members, we performed immunoprecipitation studies using antimoesin mAb and cultured baby hamster kidney (BHK) cells metabolically labeled with [35S]methionine or surface-labeled with biotin. The results indicated that moesin is directly associated with a 140-kD integral membrane protein. Using BHK cells as antigens, we obtained a mAb that recognized the 140-kD membrane protein. We next cloned a cDNA encoding the 140-kD membrane protein and identified it as CD44, a broadly distributed cell surface glycoprotein. Immunoprecipitation with various anti-CD44 mAbs showed that ezrin and radixin, as well as moesin, are associated with CD44, not only in BHK cells, but also in mouse L fibroblasts. Furthermore, immunofluorescence microscopy revealed that in both BHK and L cells, the Triton X-100-insoluble CD44 is precisely colocalized with ERM family members. We concluded that ERM family members work as molecular linkers between the cytoplasmic domain of CD44 and actin-based cytoskeletons.


Assuntos
Proteínas Sanguíneas/metabolismo , Proteínas de Transporte/metabolismo , Proteínas do Citoesqueleto , Citoesqueleto/metabolismo , Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos , Fosfoproteínas/metabolismo , Proteínas/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Retorno de Linfócitos/metabolismo , Actinas/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais , Proteínas Sanguíneas/análise , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Proteínas de Transporte/isolamento & purificação , Linhagem Celular , Cricetinae , Citoesqueleto/química , DNA Complementar/análise , Epitopos/genética , Epitopos/imunologia , Receptores de Hialuronatos , Proteínas de Membrana/análise , Camundongos , Dados de Sequência Molecular , Fosfoproteínas/análise , Testes de Precipitina , Proteínas/análise , Vírus da Raiva/química , Vírus da Raiva/imunologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/isolamento & purificação , Receptores de Retorno de Linfócitos/genética , Receptores de Retorno de Linfócitos/imunologia , Receptores de Retorno de Linfócitos/isolamento & purificação , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/imunologia , Análise de Sequência de DNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA