Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 410
Filtrar
1.
Viruses ; 16(5)2024 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-38793581

RESUMO

Rabies is a fatal encephalitic infectious disease caused by the rabies virus (RABV). RABV is highly neurotropic and replicates in neuronal cell lines in vitro. The RABV fixed strain, HEP-Flury, was produced via passaging in primary chicken embryonic fibroblast cells. HEP-Flury showed rapid adaptation when propagated in mouse neuroblastoma (MNA) cells. In this study, we compared the growth of our previously constructed recombinant HEP (rHEP) strain-based on the sequence of the HEP (HEP-Flury) strain-with that of the original HEP strain. The original HEP strain exhibited higher titer than rHEP and a single substitution at position 80 in the matrix (M) protein M(D80N) after incubation in MNA cells, which was absent in rHEP. In vivo, intracerebral inoculation of the rHEP-M(D80N) strain with this substitution resulted in enhanced viral growth in the mouse brain and a significant loss of body weight in the adult mice. The number of viral antigen-positive cells in the brains of adult mice inoculated with the rHEP-M(D80N) strain was significantly higher than that with the rHEP strain at 5 days post-inoculation. Our findings demonstrate that a single amino acid substitution in the M protein M(D80N) is associated with neurovirulence in mice owing to adaptation to mouse neuronal cells.


Assuntos
Substituição de Aminoácidos , Encéfalo , Vírus da Raiva , Raiva , Proteínas da Matriz Viral , Animais , Vírus da Raiva/genética , Vírus da Raiva/patogenicidade , Camundongos , Virulência , Encéfalo/virologia , Encéfalo/patologia , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo , Raiva/virologia , Neurônios/virologia , Neurônios/patologia , Replicação Viral , Linhagem Celular
2.
Int J Mol Sci ; 25(9)2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38731834

RESUMO

Tripartite motif (TRIM) proteins are a multifunctional E3 ubiquitin ligase family that participates in various cellular processes. Recent studies have shown that TRIM proteins play important roles in regulating host-virus interactions through specific pathways, but their involvement in response to rabies virus (RABV) infection remains poorly understood. Here, we identified that several TRIM proteins are upregulated in mouse neuroblastoma cells (NA) after infection with the rabies virus using RNA-seq sequencing. Among them, TRIM44 was found to regulate RABV replication. This is supported by the observations that downregulation of TRIM44 inhibits RABV replication, while overexpression of TRIM44 promotes RABV replication. Mechanistically, TRIM44-induced RABV replication is brought about by activating autophagy, as inhibition of autophagy with 3-MA attenuates TRIM44-induced RABV replication. Additionally, we found that inhibition of autophagy with rapamycin reverses the TRIM44-knockdown-induced decrease in LC3B expression and autophagosome formation as well as RABV replication. The results suggest that TRIM44 promotes RABV replication by an autophagy-dependent mechanism. Our work identifies TRIM44 as a key host factor for RABV replication, and targeting TRIM44 expression may represent an effective therapeutic strategy.


Assuntos
Autofagia , Vírus da Raiva , Proteínas com Motivo Tripartido , Replicação Viral , Autofagia/genética , Animais , Camundongos , Proteínas com Motivo Tripartido/metabolismo , Proteínas com Motivo Tripartido/genética , Vírus da Raiva/fisiologia , Vírus da Raiva/genética , Linhagem Celular Tumoral , Humanos , Raiva/virologia , Raiva/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Interações Hospedeiro-Patógeno
3.
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 , Vírus da Raiva/imunologia , Vírus da Raiva/genética , Vírus da Raiva/patogenicidade , Proteína DEAD-box 58/metabolismo , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/imunologia , Animais , Humanos , Raiva/imunologia , Raiva/virologia , RNA Viral/genética , Receptores Imunológicos/metabolismo , Vacina Antirrábica/imunologia , Linhagem Celular , Transdução de Sinais , Camundongos , Replicação Viral , Interferon Tipo I/metabolismo , Interferon Tipo I/imunologia
4.
BMC Neurosci ; 25(1): 9, 2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38383317

RESUMO

BACKGROUND: A pseudotyped modified rabies virus lacking the rabies glycoprotein (G-protein), which is crucial for transsynaptic spread, can be used for monosynaptic retrograde tracing. By coupling the pseudotyped virus with transgene expression of the G-protein and the avian leukosis and sarcoma virus subgroup A receptor (TVA), which is necessary for cell entry of the virus, researchers can investigate specific neuronal populations. Responder mouse lines, like the RΦGT mouse line, carry the genes encoding the G-protein and TVA under Cre-dependent expression. These mouse lines are valuable tools because they reduce the number of viral injections needed compared to when using helper viruses. Since RΦGT mice do not express Cre themselves, introducing the pseudotyped rabies virus into their brain should not result in viral cell entry or spread. RESULTS: We present a straightforward flowchart for adequate controls in tracing experiments, which we employed to demonstrate Cre-independent expression of TVA in RΦGT mice. CONCLUSIONS: Our observations revealed TVA leakage, indicating that RΦGT mice should be used with caution for transgene expression of TVA. Inaccurate tracing outcomes may occur if TVA is expressed in the absence of Cre since background leakage leads to nonspecific cell entry. Moreover, conducting appropriate control experiments can identify the source of potential caveats in virus-based neuronal tracing experiments.


Assuntos
Proteínas Aviárias , Vírus da Raiva , Camundongos , Animais , Design de Software , Receptores Virais/genética , Receptores Virais/metabolismo , Proteínas Aviárias/metabolismo , Vírus da Raiva/genética , Vírus da Raiva/metabolismo , Proteínas de Ligação ao GTP/metabolismo
5.
mBio ; 15(3): e0288023, 2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38349129

RESUMO

Infection with neurotropic viruses may result in changes in host behavior, which are closely associated with degenerative changes in neurons. The lyssavirus genus comprises highly neurotropic viruses, including the rabies virus (RABV), which has been shown to induce degenerative changes in neurons, marked by the self-destruction of axons. The underlying mechanism by which the RABV degrades neuronal cytoskeletal proteins remains incomplete. In this study, we show that infection with RABV or overexpression of its M protein can disrupt mitochondrial metabolism by binding to Slc25a4. This leads to a reduction in NAD+ production and a subsequent influx of Ca2+ from the endoplasmic reticulum and mitochondria into the cytoplasm of neuronal cell lines, activating Ca2+-dependent proteinase calpains that degrade α-tubulin. We further screened the M proteins of different lyssaviruses and discovered that the M protein of the dog-derived RABV strain (DRV) does not degrade α-tubulin. Sequence analysis of the DRV M protein and that of the lab-attenuated RABV strain CVS revealed that the 57th amino acid is vital for M-induced microtubule degradation. We generated a recombinant RABV with a mutation at the 57th amino acid position in its M protein and showed that this mutation reduces α-tubulin degradation in vitro and axonal degeneration in vivo. This study elucidates the mechanism by which lyssavirus induces neuron degeneration.IMPORTANCEPrevious studies have suggested that RABV (rabies virus, the representative of lyssavirus) infection induces structural abnormalities in neurons. But there are few articles on the mechanism of lyssavirus' effect on neurons, and the mechanism of how RABV infection induces neurological dysfunction remains incomplete. The M protein of lyssavirus can downregulate cellular ATP levels by interacting with Slc25a4, and this decrease in ATP leads to a decrease in the level of NAD+ in the cytosol, which results in the release of Ca2+ from the intracellular calcium pool, the endoplasmic reticulum, and mitochondria. The presence of large amounts of Ca2+ in the cytoplasm activates Ca2+-dependent proteases and degrades microtubule proteins. The amino acid 57 of M protein is the key site determining its disruption of mitochondrial metabolism and subsequent neuron degeneration.


Assuntos
Lyssavirus , Vírus da Raiva , Raiva , Animais , Cães , Lyssavirus/genética , Tubulina (Proteína)/metabolismo , NAD/metabolismo , Vírus da Raiva/genética , Vírus da Raiva/metabolismo , Raiva/metabolismo , Neurônios , Microtúbulos/metabolismo , Mitocôndrias/metabolismo , Aminoácidos/metabolismo , Degeneração Neural/metabolismo , Trifosfato de Adenosina/metabolismo
6.
Emerg Microbes Infect ; 13(1): 2300461, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38164714

RESUMO

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.


Assuntos
Doenças do Gato , Doenças do Cão , Vacina Antirrábica , Vírus da Raiva , Gatos , Cães , Humanos , Animais , Camundongos , Vírus da Raiva/genética , Vacina Antirrábica/genética , Imunidade Humoral , Anticorpos Antivirais , Adenoviridae/genética
7.
Int J Biol Macromol ; 255: 128085, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37977454

RESUMO

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.


Assuntos
Vacina Antirrábica , Vírus da Raiva , Raiva , Vacinas de DNA , Humanos , Animais , Camundongos , Raiva/prevenção & controle , Escherichia coli , Anticorpos Antivirais , Vacina Antirrábica/genética , Vírus da Raiva/genética , Epitopos de Linfócito B/genética
8.
Vet. zootec ; 31: 1-7, 2024.
Artigo em Inglês | LILACS, VETINDEX | ID: biblio-1552662

RESUMO

Rabies is a fatal zoonotic disease that affects several mammals. Hematophagous bats are recognized hosts of the rabies virus, and their main food source is the blood of other mammals, particularly cattle. During feeding, bats transmit the virus to cattle, which are victims of the disease, contributing to economic losses and increasing the risk of infection for humans. Based on this affinity in the rabies cycle between bats and cattle, the objective of this study was to analyze the phylogenetic relationships of rabies virus samples in cattle and bats. The G gene of the rabies virus was chosen for this study because it is directly related to the infection process. Nucleotide sequences of the viral G gene were selected from GenBank for samples obtained from infected cattle and bats. Maximum parsimony analyses were conducted using the Molecular Evolutionary Genetics Analysis software. The Maxima Parsimony tree indicated a phylogenetic relationship between the G genes of both hosts, indicating that the virus evolved from bats to cattle. Analysis of parsimoniously informative sites revealed that the viral G gene carried specific mutations in each host. Knowledge of the evolutionary relationships between the rabies virus and its hosts is critical for identifying potential new hosts and the possible routes of infection for humans.


A Raiva é uma zoonose fatal que infecta várias espécies de mamíferos. Os morcegos hematófagos são reconhecidos como hospedeiros do vírus da Raiva e sua principal fonte de alimento é o sangue de outros mamíferos, especialmente os bovinos. Quando se alimentam, os morcegos transmitem o vírus para o bovino os quais são vítimas da doença, contribuindo para perdas econômicas e riscos de infecção para humanos. Baseado nesta afinidade do ciclo da Raiva entre morcegos e bovinos, o objetivo deste estudo foi analisar as relações filogenéticas de amostras do vírus da Raiva em ambos os hospedeiros, bovinos e morcegos. O gene G do vírus da Raiva foi escolhido para esta pesquisa porque ele está diretamente relacionado ao processo de infecção. Sequências de nucleotídeos do gene G viral foram selecionadas no GenBank a partir de amostras obtidas de bovinos e morcegos infectados. Análises de Máxima Parcimônia foram conduzidas utilizando o software Molecular Evolutionary Genetics Analysis. A árvore de Máxima Parcimônia indicou uma relação filogenética entre o gene G de ambos os hospedeiros, indicando que o vírus evoluiu dos morcegos para os bovinos. A análise dos sítios parcimoniosamente informativos revelou que o gene G viral apresentou mutações específicas em cada hospedeiro. O conhecimento sobre as relações evolutivas do vírus da Raiva e seus hospedeiros é crucial para identificar nos hospedeiros potenciais e novas rotas possíveis de infecção para humanos.


La rabia es una zoonosis fatal que infecta a varias especies de mamíferos. Los murciélagos hematófagos son reconocidos como huéspedes del virus de la rabia y su principal fuente de alimentación es la sangre de otros mamíferos, especialmente del ganado. Al alimentarse, los murciélagos transmiten el virus al ganado que es víctima de la enfermedad, contribuyendo a pérdidas económicas y riesgos de infección para los humanos. Basado en esta afinidad del ciclo de la rabia entre murciélagos y ganado, el objetivo de este estudio fue analizar las relaciones filogenéticas de las muestras de virus de la rabia tanto en huéspedes, ganado y murciélagos. El gen G del virus de la rabia fue elegido para esta investigación porque está directamente relacionado con el proceso de infección. Las secuencias de nucleótidos del gen G viral se seleccionaron en GenBank a partir de muestras obtenidas de bovinos y murciélagos infectados. Los análisis de parsimonia máxima se realizaron utilizando el software Molecular Evolutionary Genetics Analysis. El árbol de Máxima Parsimônia indicó una relación filogenética entre el gen G de ambos huéspedes, indicando que el virus evolucionó de murciélagos a bovinos. El análisis de los sitios parsimoniosamente informativos reveló que el gen G viral presentaba mutaciones específicas en cada huésped. El conocimiento sobre las relaciones evolutivas del virus de la rabia y sus huéspedes es crucial para identificar huéspedes potenciales y nuevas posibles rutas de infección para humanos.


Assuntos
Animais , Filogenia , Vírus da Raiva/genética , Viroses/veterinária , Quirópteros/virologia
9.
Int J Med Sci ; 20(10): 1282-1292, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37786447

RESUMO

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.


Assuntos
Vacina Antirrábica , Vírus da Raiva , Raiva , Chlorocebus aethiops , Humanos , Animais , Camundongos , Suínos , Vírus da Raiva/genética , Raiva/prevenção & controle , Células HEK293 , Células Vero , Anticorpos Antivirais , Glicoproteínas/genética , Vacina Antirrábica/genética , Proteínas do Envelope Viral/genética , Proteínas Recombinantes
10.
J Med Virol ; 95(9): e29110, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37728394

RESUMO

Rabies, a lethal zoonotic encephalitis, remains a significant global health concern, causing an estimated 60 000 annual fatalities worldwide. Dogs serve as the primary reservoirs and vectors for transmitting this infection to humans. Definitive diagnosis of rabies in both human and animal cases necessitates laboratory testing involving various clinical specimens. However, the complexity of laboratory infrastructure and the need for skilled personnel, along with the challenge of maintaining cold-chain integrity during sample referral, hinder the decentralization of diagnostic facilities. This study aimed to assess the efficacy of the Truenat rabies assay, a rapid, portable, semiautomated, and closed PCR-based system, for the diagnosis of rabies in both humans and animals. The Truenat assay demonstrated a sensitivity of 100% and a specificity of 86.96% when compared with the fluorescent antibody test (FAT), as the reference standard, on 147 canine brain samples tested. Notably, the Truenat assay exhibited a sensitivity and specificity of 100% when tested on 48 human brain specimens. Furthermore, an examination of 148 human antemortem samples (cerebrospinal fluid, saliva, and skin biopsy) using both the Truenat assay and a validated real-time reverse transcriptase PCR assay revealed a κ value of 0.505, indicative of a moderate level of agreement between the two tests. Thus, the Truenat assay offers a robust, reliable, and affordable point-of-care solution to enhance rabies diagnostic capacity in endemic areas.


Assuntos
Vírus da Raiva , Raiva , Humanos , Cães , Animais , Vírus da Raiva/genética , Raiva/diagnóstico , Raiva/veterinária , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Bioensaio , Biópsia
11.
Appl Microbiol Biotechnol ; 107(12): 3955-3966, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37166480

RESUMO

Fusion tag technology is an important tool for rapid separation, purification, and characterization of proteins. Combined with monoclonal antibodies, tag epitope systems can be rapidly adapted to many assay systems. A monoclonal antibody that reacts with the matrix protein of the rabies virus CVS-11 strain was reported. The epitope (termed M) targeted by this antibody contains only six amino acids. We examine whether this specific sequence epitope can be applied as a protein tag. We show ectopic expression of M-tagged proteins has little impact on cell viability or major signaling pathways. The M tag system can be used for western blotting, immunoprecipitation, immunofluorescence staining, and flow cytometry assays. The results indicate the specificity, sensitivity, and versatility of this novel epitope tag system are comparable to the widely used FLAG tag system, providing researchers with an additional tool for molecular analysis. KEY POINTS: • A short peptide (Pro Pro Tyr Asp Asp Asp) can be applied as a new tag. • The new epitope-tagging fusion system has no effect on the main cellular signaling pathway. • The epitope-tagging fusion system can be widely used for western blotting, immunoprecipitation, immunofluorescence, flow cytometry, etc.


Assuntos
Vírus da Raiva , Epitopos , Vírus da Raiva/genética , Peptídeos/metabolismo , Anticorpos Monoclonais , Western Blotting
12.
Front Cell Infect Microbiol ; 13: 1013842, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36798087

RESUMO

Macrophages are amongst the first immune cells that encounter rabies virus (RABV) at virus entry sites. Activation of macrophages is essential for the onset of a potent immune response, but insights into the effects of RABV on macrophage activation are scarce. In this study we performed high-throughput sequencing on RNA extracted from macrophages that were exposed to RABV for 48 hours, and compared their transcriptional profiles to that of non-polarized macrophages (M0), and macrophages polarized towards the canonical M1, M2a and M2c phenotypes. Our analysis revealed that RABV-stimulated macrophages show high expression of several M1, M2a and M2c signature genes. Apart from their partial resemblance to these phenotypes, unbiased clustering analysis revealed that RABV induces a unique and distinct polarization program. Closer examination revealed that RABV induced multiple pathways related to the interferon- and antiviral response, which were not induced under other classical polarization strategies. Surprisingly, our data show that RABV induces an activated rather than a fully suppressed macrophage phenotype, triggering virus-induced activation and polarization. This includes multiple genes with known antiviral (e.g. APOBEC3A, IFIT/OAS/TRIM genes), which may play a role in anti-RABV immunity.


Assuntos
Vírus da Raiva , Raiva , Humanos , Vírus da Raiva/genética , Transcriptoma , Macrófagos/metabolismo , Antivirais/farmacologia
13.
Emerg Microbes Infect ; 11(1): 1439-1451, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35579916

RESUMO

Three or four intramuscular doses of the inactivated human rabies virus vaccines are needed for pre- or post-exposure prophylaxis in humans. This procedure has made a great contribution to prevent human rabies deaths, which bring huge economic burdens in developing countries. Herein, a recombinant adeno-associated virus serotype 9, AAV9-RABVG, harbouring a RABV G gene, was generated to serve as a single dose rabies vaccine candidate. The RABV G protein was stably expressed in the 293T cells infected with AAV9-RABVG. A single dose of 2 × 1011 v.p. of AAV9-RABVG induced robust and long-term positive seroconversions in BALB/c mice with a 100% survival from a lethal RABV challenge. In Cynomolgus Macaques vaccinated with a single dose of 1 × 1013 v.p. of AAV9-RABVG, the titres of rabies VNAs increased remarkably from 2 weeks after immunity, and maintained over 31.525 IU/ml at 52 weeks. More DCs were activated significantly for efficient antigen presentations of RABV G protein, and more B cells were activated to be responsible for antibody responses. Significantly more RABV G specific IFN-γ-secreting CD4+ and CD8+ T cells, and IL-4-secreting CD4+ T cells were activated, and significantly higher levels of IL-2, IFN-γ, IL-4, and IL-10 were secreted to aid immune responses. Overall, the AAV9-RABVG was a single dose rabies vaccine candidate with great promising by inducing robust, long-term humoral responses and both Th1 and Th2 cell-mediated immune responses in mice and non-human primates.


Assuntos
Vacina Antirrábica , Vírus da Raiva , Raiva , Animais , Anticorpos Antivirais , Dependovirus/genética , Proteínas de Ligação ao GTP/genética , Imunidade Celular , Interleucina-4/genética , Camundongos , Camundongos Endogâmicos BALB C , Primatas , Raiva/prevenção & controle , Vírus da Raiva/genética , Sorogrupo
14.
Vopr Virusol ; 67(2): 107-114, 2022 05 05.
Artigo em Russo | MEDLINE | ID: mdl-35521983

RESUMO

Rabies is almost ubiquitous (except in certain areas) and poses a significant danger to both animals and humans. Every year around 55,000 people die from this disease worldwide. In the Russian Federation alone 400,000- 450,000 patients annually apply for anti-rabies treatment. In the absolute majority of cases human infection is caused by contact with infected animals. In RF, a number of cultured inactivated anti-rabies vaccines for medical and veterinary purposes have been developed, registered and used for specific prevention of rabies. These vaccine preparations have shown high effectiveness in preventing infection in domestic and farm animals. At the same time, the main reservoir of the rabies virus (Mononegavirales: Rhabdoviridae: Lyssavirus) (RV) are wild carnivores (Mammalia: Carnivora). For the purpose of their oral immunization, live virus vaccines from attenuated (fixed) strains of RV that are little resistant in the external environment are used. In Western Europe and North America there is successful experience with recombinant anti-rabies vaccine preparations containing a viral glycoprotein gene (G-protein). Such vaccines are safe for humans and animals. In Russia also had been developed a vector anti-rabies vaccine based on adenovirus (Adenoviridae), which can be used to combat this infection. Currently, in addition to classical rabies, diseases caused by new, previously unknown lyssaviruses (Lyssavirus) are becoming increasingly important. Bats (Mammalia: Microchiroptera) are their vectors. Cases of illness and death after contact with these animals have been described. In the near future, we should expect the development of new vaccines that will provide protection not only against RV, but also against other lyssaviruses.


Assuntos
Quirópteros , Lyssavirus , Vacina Antirrábica , Vírus da Raiva , Raiva , Animais , Humanos , Lyssavirus/genética , Raiva/epidemiologia , Raiva/prevenção & controle , Vírus da Raiva/genética , Vacinas Sintéticas
15.
J Virol ; 96(9): e0011122, 2022 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-35404083

RESUMO

The rabies virus (RABV) phosphoprotein (P protein) is expressed as several isoforms, which differ in nucleocytoplasmic localization and microtubule (MT) association, mediated by several sequences, including nuclear localization (NLS) and export (NES) sequences. This appears to underpin a functional diversity enabling multiple functions in viral replication and modulation of host biology. Mechanisms regulating trafficking are poorly defined, but phosphorylation by protein kinase C (PKC) in the P protein C-terminal domain (PCTD) regulates nuclear trafficking, mediated by PCTD-localized NLS/NES sequences, indicating that phosphorylation contributes to functional diversity. The molecular mechanism underlying the effects of PKC, and potential roles in regulating other host-cell interactions are unresolved. Here, we assess effects of phosphorylation on the P3 isoform, which differs from longer isoforms through an ability to localize to the nucleus and associate with MTs, which are associated with antagonism of interferon (IFN) signaling. We find that phosphomimetic mutation of the PKC site S210 inhibits nuclear accumulation and MT association/bundling. Structural analysis indicated that phosphomimetic mutation induces no significant structural change to the NLS/NES but results in the side chain of N226 switching its interactions from E228, within the NES, to E210. Intriguingly, N226 is the sole substituted residue between the PCTD of the pathogenic IFN-resistant RABV strain Nishigahara and a derivative attenuated IFN-sensitive strain Ni-CE, inhibiting P3 nuclear localization and MT association. Thus, S210 phosphorylation appears to impact on N226/E228 to regulate P protein localization, with N226 mutation in Ni-CE mimicking a constitutively phosphorylated state resulting in IFN sensitivity and attenuation. IMPORTANCE Rabies virus P protein is a multifunctional protein with critical roles in replication and manipulation of host-cell processes, including subversion of immunity. This functional diversity involves interactions of several P protein isoforms with the cell nucleus and microtubules. Previous studies showed that phosphorylation of the P protein C-terminal domain (PCTD) at S210, near nuclear trafficking sequences, regulates nucleocytoplasmic localization, indicating key roles in functional diversity. The molecular mechanisms of this regulation have remained unknown. Here, we show that phosphomimetic mutation of S210 regulates nuclear localization and MT association. This regulation does not appear to result from disrupted PCTD structure, but rather from a switch of specific side chain interactions of N226. Intriguingly, N226 was previously implicated in P protein nuclear localization/MT association, immune evasion, and RABV pathogenesis, through undefined mechanisms. Our data indicate that the S210-N226 interface is a key regulator of virus-host interactions, which is significant for pathogenesis.


Assuntos
Chaperonas Moleculares , Vírus da Raiva , Proteínas Estruturais Virais , Animais , Núcleo Celular/metabolismo , Fosforilação , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Vírus da Raiva/genética , Vírus da Raiva/metabolismo
16.
Front Immunol ; 13: 823949, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35173733

RESUMO

Middle East respiratory syndrome coronavirus (MERS-CoV) is an emergent coronavirus that has caused frequent zoonotic events through camel-to-human spillover. An effective camelid vaccination strategy is probably the best way to reduce human exposure risk. Here, we constructed and evaluated an inactivated rabies virus-vectored MERS-CoV vaccine in mice, camels, and alpacas. Potent antigen-specific antibody and CD8+ T-cell responses were generated in mice; moreover, the vaccination reduced viral replication and accelerated virus clearance in MERS-CoV-infected mice. Besides, protective antibody responses against both MERS-CoV and rabies virus were induced in camels and alpacas. Satisfyingly, the immune sera showed broad cross-neutralizing activity against the three main MERS-CoV clades. For further characterization of the antibody response induced in camelids, MERS-CoV-specific variable domains of heavy-chain-only antibody (VHHs) were isolated from immunized alpacas and showed potent prophylactic and therapeutic efficacies in the Ad5-hDPP4-transduced mouse model. These results highlight the inactivated rabies virus-vectored MERS-CoV vaccine as a promising camelid candidate vaccine.


Assuntos
Camelídeos Americanos/virologia , Camelus/virologia , Infecções por Coronavirus/veterinária , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Linfócitos T CD8-Positivos/imunologia , Camelídeos Americanos/imunologia , Camelus/imunologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Cricetinae , Feminino , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vírus da Raiva/genética , Vírus da Raiva/imunologia , Vacinação , Vacinas Sintéticas/imunologia , Células Vero , Vacinas Virais/genética
18.
Transbound Emerg Dis ; 69(4): e254-e266, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34403194

RESUMO

Cystic echinococcosis (CE), caused by Echinococcus granulosus (E, is a zoonosis with a worldwide distribution, resulting in heavy impact to public health and social economics. In this study, we generated a recombinant rabies virus (RABV) expressing EG95 protein of E. granulosus (LBNSE-EG95) as a bivalent candidate vaccine for use in sheep and cattle against CE and rabies, which is another severe health threat in CE-endemic areas. It was found that EG95 was successfully expressed without altering the pathogenicity of parent LBNSE vector. Further study showed that LBNSE-EG95 immunization in mice elicited activation of dendric cells (DCs) and B cells and induced Th1-/Th2-mediated cellular immune responses, leading to robust production of RABV neutralizing antibodies and high level of EG95-sepecific antibodies with more than 90% protection against CE. In addition, single dose of LBNSE-EG95 conferred full protection against lethal RABV challenge in mice. Collectively, these results suggest that the recombinant LBNSE-EG95 has the potential to be developed as an efficient bivalent vaccine for sheep and cattle use in endemic areas of CE and rabies.


Assuntos
Doenças dos Bovinos , Equinococose , Echinococcus granulosus , Orthopoxvirus , Vacina Antirrábica , Vírus da Raiva , Raiva , Doenças dos Roedores , Doenças dos Ovinos , Animais , Anticorpos Antivirais , Bovinos , Equinococose/prevenção & controle , Equinococose/veterinária , Echinococcus granulosus/genética , Camundongos , Raiva/prevenção & controle , Raiva/veterinária , Vírus da Raiva/genética , Proteínas Recombinantes/genética , Ovinos , Doenças dos Ovinos/prevenção & controle
19.
Viruses ; 13(11)2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34835093

RESUMO

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.


Assuntos
Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Raiva/prevenção & controle , Proteínas Estruturais Virais/imunologia , Animais , Humanos , Raiva/imunologia , Raiva/virologia , Vacina Antirrábica/genética , Vírus da Raiva/genética , Genética Reversa/métodos , Vacinas Atenuadas , Proteínas Estruturais Virais/genética , Replicação Viral
20.
Viruses ; 13(10)2021 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-34696419

RESUMO

Molecular details of field rabies virus (RABV) adaptation to cell culture replication are insufficiently understood. A better understanding of adaptation may not only reveal requirements for efficient RABV replication in cell lines, but may also provide novel insights into RABV biology and adaptation-related loss of virulence and pathogenicity. Using two recombinant field rabies virus clones (rRABV Dog and rRABV Fox), we performed virus passages in three different cell lines to identify cell culture adaptive mutations. Ten passages were sufficient for the acquisition of adaptive mutations in the glycoprotein G and in the C-terminus of phosphoprotein P. Apart from the insertion of a glycosylation sequon via the mutation D247N in either virus, both acquired additional and cell line-specific mutations after passages on BHK (K425N) and MDCK-II (R346S or R350G) cells. As determined by virus replication kinetics, complementation, and immunofluorescence analysis, the major bottleneck in cell culture replication was the intracellular accumulation of field virus G protein, which was overcome after the acquisition of the adaptive mutations. Our data indicate that limited release of extracellular infectious virus at the plasma membrane is a defined characteristic of highly virulent field rabies viruses and we hypothesize that the observed suboptimal release of infectious virions is due to the inverse correlation of virus release and virulence in vivo.


Assuntos
Antígenos Virais/genética , Vírus da Raiva/genética , Proteínas do Envelope Viral/genética , Liberação de Vírus/genética , Animais , Anticorpos Antivirais/sangue , Antígenos Virais/metabolismo , Técnicas de Cultura de Células , Linhagem Celular , Cães , Glicoproteínas/genética , Glicosilação , Mutação Puntual/genética , Raiva/virologia , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/genética , Vírion/metabolismo , Virulência/genética , Replicação Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA