Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 176
Filtrar
1.
PLoS Negl Trop Dis ; 16(1): e0010087, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35051178

RESUMO

INTRODUCTION: Globally, traditional medicine is widely used to treat a variety of injuries and illnesses, including dog bites, and exposures that are risky for rabies. However, efficacy of most traditional remedies used for rabies prevention or treatment has not been demonstrated in controlled trials or proven in community-based surveys. METHODS: Six databases were searched including the terms rabies, traditional treatment, traditional remedy, traditional therapy, traditional medicine, and medicinal treatment to review traditional remedies used in the prevention and treatment of rabies. In addition, published literature of rabies transmission dynamics was used to estimate statistical likelihood of dog bite victims developing rabies to provide clarity as to why traditional healers have a high apparent success rate when preventing death from rabies in victims bitten by suspected rabid dogs. RESULTS: Literature review yielded 50 articles, including three controlled experiments, that described use of traditional remedies for rabies prevention and treatment. Traditional remedies for rabies ranged from plant- or animal-based products to spiritual rituals; however, only a few controlled mice trials were conducted, and none of these trials demonstrated efficacy in preventing or treating rabies. Risk of dying from rabies after a bite from a dog with unknown rabies status is low, 1.90% (0.05%-29.60%). Therefore, traditional healers had a 98.10% (70.40%-99.95%) apparent success rate in preventing death from suspected rabid dog bites despite inefficaciousness of herbal remedies. CONCLUSION: There was no universal plant species or route of administration that was consistently used for rabies prevention or treatment across countries. No traditional remedy was efficacious in the prevention or treatment of rabies in randomized controlled experiments. Understanding the cultural context under which traditional remedies are used may facilitate collaboration of traditional healers with the modern medical system to ensure timely and appropriate use of proven therapies for prevention and clinical management of rabies.


Assuntos
Doenças do Cão/transmissão , Medicina Tradicional/métodos , Fitoterapia/métodos , Profilaxia Pós-Exposição/métodos , Raiva/prevenção & controle , Animais , Doenças do Cão/virologia , Cães , Raiva/tratamento farmacológico , Vírus da Raiva/efeitos dos fármacos
2.
J Virol ; 96(2): e0147321, 2022 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-34757839

RESUMO

Rabies is an old zoonotic disease caused by rabies virus (RABV), but the pathogenic mechanism of RABV is still not completely understood. Lipid droplets (LDs) have been reported to play a role in pathogenesis of several viruses. However, their role in RABV infection remains unclear. Here, we initially found that RABV infection upregulated LD production in multiple cells and mouse brains. After treatment with atorvastatin, a specific inhibitor of LDs, RABV replication in N2a cells decreased. Then we found that RABV infection could upregulate N-myc downstream regulated gene-1 (NDRG1), which in turn enhanced the expression of diacylglycerol acyltransferase 1/2 (DGAT1/2). DGAT1/2 could elevate cellular triglyceride synthesis and ultimately promote intracellular LD formation. Furthermore, we found that RABV-M and RABV-G, which were mainly involved in the viral budding process, could colocalize with LDs, indicating that RABV might utilize LDs as a carrier to facilitate viral budding and eventually increase virus production. Taken together, our study reveals that lipid droplets are beneficial for RABV replication, and their biogenesis is regulated via the NDRG1-DGAT1/2 pathway, which provides novel potential targets for developing anti-RABV drugs. IMPORTANCE Lipid droplets have been proven to play an important role in viral infections, but their role in RABV infection has not yet been elaborated. Here, we find that RABV infection upregulates the generation of LDs by enhancing the expression of N-myc downstream regulated gene-1 (NDRG1). Then NDRG1 elevated cellular triglycerides synthesis by increasing the activity of diacylglycerol acyltransferase 1/2 (DGAT1/2), which promotes the biogenesis of LDs. RABV-M and RABV-G, which are the major proteins involved in viral budding, could utilize LDs as a carrier for transport to cell membrane, resulting in enhanced virus budding. Our findings will extend the knowledge of lipid metabolism in RABV infection and help to explore potential therapeutic targets for RABV.


Assuntos
Gotículas Lipídicas/metabolismo , Vírus da Raiva/fisiologia , Raiva/virologia , Liberação de Vírus , Replicação Viral , Animais , Anticolesterolemiantes/farmacologia , Atorvastatina/farmacologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diacilglicerol O-Aciltransferase/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Gotículas Lipídicas/efeitos dos fármacos , Camundongos , Neurônios/metabolismo , Neurônios/virologia , Raiva/metabolismo , Vírus da Raiva/efeitos dos fármacos , Triglicerídeos/metabolismo , Proteínas Estruturais Virais/metabolismo , Liberação de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
3.
Vet Microbiol ; 262: 109241, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34555731

RESUMO

Rabies, caused by rabies virus (RABV), is one of the most important neurotropic zoonoses and poses a severe threat to human and animal health. Exploration of its mechanism of neural transmission is meaningful but still insufficient. Here, we described the effects of microtubule-depolymerizing drugs and inhibitors of microtubule motor proteins on RABV infection. Colchicine, a microtubule-depolymerizing drug, significantly impeded RABV production in N2a cells. Overexpression of CC1 or p50 attenuated viral infection through the functional disruption of cytoplasmic dynein, which was consistent with the inhibitory effect of Na3VO4, a dynein activity inhibitor. Moreover, transfection with Flag-KHCct impaired RABV infection, as cytoplasmic kinesin-based motility was blocked. These results demonstrated that RABV can infect N2a cells in a manner that depends on microtubule integrity as well as dynein and kinesin function.


Assuntos
Dineínas , Cinesinas , Vírus da Raiva , Raiva , Animais , Antivirais/farmacologia , Dineínas/metabolismo , Cinesinas/metabolismo , Microtúbulos/metabolismo , Raiva/veterinária , Raiva/virologia , Vírus da Raiva/efeitos dos fármacos
4.
Artigo em Inglês | MEDLINE | ID: mdl-33852711

RESUMO

Currently, the Milwaukee protocol presents healing results in human beings affected by the rabies virus. However, there are many points to clarify on the action of drugs and the immune mechanism involved in the evolution of the disease. One of the drugs used is biopterin, which is an important cofactor for nitric oxide, important for preventing vasospasm. Thus, we describe the effect of biopterin on some inflammatory factors in a rabies virus infection developed in an animal model. The immunological mediators studied in animals infected with rabies virus submitted to doses of sapropterin were Anti-RABV, IL-6, IL-2, IL-17a, INF-gamma and Anti-iNOS. It is suggested that the medication in the context of a RABV infection already installed, had the effect of modulating the inflammatory mechanisms mainly linked to the permeability of the blood-brain barrier and the migration of cytotoxic cells.


Assuntos
Biopterinas/análogos & derivados , Biopterinas/farmacologia , Vírus da Raiva/efeitos dos fármacos , Raiva/tratamento farmacológico , Animais , Encéfalo , Sistema Nervoso Central , Modelos Animais de Doenças , Humanos , Camundongos , Vírus da Raiva/isolamento & purificação
5.
Antiviral Res ; 188: 105016, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33444703

RESUMO

ABMA and its analogue DABMA are two molecules of the adamantane family known to perturbate the endosomal pathway and to inhibit cell infection of several RNA and DNA viruses. Their activity against Rabies Virus (RABV) infection has already been demonstrated in vitro. (Wu et al., 2017, 2019). Here, we describe in more details their mechanism of action by comparison to Arbidol (umifenovir) and Ribavirin, two broad spectrum antivirals against emerging viruses such as Lassa, Ebola, influenza and Hantaan viruses. ABMA and DABMA, delivered 2 h pre-infection, inhibit RABV infection in vitro with an EC50 of 7.8 µM and 14 µM, respectively. They act at post-entry, by causing RABV accumulation within the endosomal compartment and DABMA specifically diminishes the expression of the GTPase Rab7a controlling the fusion of early endosomes to late endosomes or lysosomes. This may suggest that ABMA and DABMA act at different stages of the late endosomal pathway as supported by their different profile of synergy/antagonism with the fusion inhibitor Arbidol. This difference is further confirmed by the RABV mutants induced by successive passages under increasing selective pressure showing a particular involvement of the viral G protein in the DABMA inhibition while ABMA inhibition induces less mutations dispersed in the M, G and L viral proteins. These results suggest new therapeutic perspectives against rabies.


Assuntos
Adamantano/farmacologia , Antivirais/farmacologia , Benzilaminas/farmacologia , Vírus da Raiva/efeitos dos fármacos , Animais , Linhagem Celular , Farmacorresistência Viral , Sinergismo Farmacológico , Endossomos/metabolismo , Indóis/farmacologia , Mutação , Vírus da Raiva/genética , Vírus da Raiva/fisiologia , Ribavirina/farmacologia , Proteínas Virais/genética , Internalização do Vírus/efeitos dos fármacos
6.
Mol Ther ; 29(3): 1174-1185, 2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33352107

RESUMO

Self-amplifying RNA (saRNA) is a cutting-edge platform for both nucleic acid vaccines and therapeutics. saRNA is self-adjuvanting, as it activates types I and III interferon (IFN), which enhances the immunogenicity of RNA vaccines but can also lead to inhibition of translation. In this study, we screened a library of saRNA constructs with cis-encoded innate inhibiting proteins (IIPs) and determined the effect on protein expression and immunogenicity. We observed that the PIV-5 V and Middle East respiratory syndrome coronavirus (MERS-CoV) ORF4a proteins enhance protein expression 100- to 500-fold in vitro in IFN-competent HeLa and MRC5 cells. We found that the MERS-CoV ORF4a protein partially abates dose nonlinearity in vivo, and that ruxolitinib, a potent Janus kinase (JAK)/signal transducer and activator of transcription (STAT) inhibitor, but not the IIPs, enhances protein expression of saRNA in vivo. Both the PIV-5 V and MERS-CoV ORF4a proteins were found to enhance the percentage of resident cells in human skin explants expressing saRNA and completely rescued dose nonlinearity of saRNA. Finally, we observed that the MERS-CoV ORF4a increased the rabies virus (RABV)-specific immunoglobulin G (IgG) titer and neutralization half-maximal inhibitory concentration (IC50) by ∼10-fold in rabbits, but not in mice or rats. These experiments provide a proof of concept that IIPs can be directly encoded into saRNA vectors and effectively abate the nonlinear dose dependency and enhance immunogenicity.


Assuntos
Imunidade Inata/efeitos dos fármacos , Imunogenicidade da Vacina , Biossíntese de Proteínas/efeitos dos fármacos , Vacinas Sintéticas/farmacologia , Proteínas do Envelope Viral/administração & dosagem , Animais , Linhagem Celular , Vírus da Encefalite Equina Venezuelana/efeitos dos fármacos , Vírus da Encefalite Equina Venezuelana/imunologia , Vírus da Encefalite Equina Venezuelana/patogenicidade , Fibroblastos , Regulação da Expressão Gênica , Células HeLa , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunoglobulina G/biossíntese , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Janus Quinases/antagonistas & inibidores , Janus Quinases/genética , Janus Quinases/imunologia , Camundongos , Coronavírus da Síndrome Respiratória do Oriente Médio/efeitos dos fármacos , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Coronavírus da Síndrome Respiratória do Oriente Médio/patogenicidade , NF-kappa B/genética , NF-kappa B/imunologia , Nitrilas , Vírus da Parainfluenza 5/efeitos dos fármacos , Vírus da Parainfluenza 5/imunologia , Vírus da Parainfluenza 5/patogenicidade , Pirazóis/farmacologia , Pirimidinas , Coelhos , Vírus da Raiva/efeitos dos fármacos , Vírus da Raiva/imunologia , Vírus da Raiva/patogenicidade , Ratos , Fatores de Transcrição STAT/antagonistas & inibidores , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/imunologia , Transdução de Sinais , Vacinas Sintéticas/biossíntese , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas de mRNA
7.
J Neurovirol ; 26(5): 764-768, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32725420

RESUMO

Rabies is a neurological disease with 100% lethality. Some of the rare human patients who survived after multiple drug treatment had severe sequelae. The present study showed that after 48 h of RABV inoculation, mice injected intracerebrally with anti-RABV F (ab')2 plus Bioporter® showed 70% survival compared to the control group, suggesting that transfection of anti-RABV antibodies to the brain may prevent or delay the spread of RABV at an early stage of infection. This result may provide important protocol results in intracellular antibody delivery to prevent the fatal outcome of the disease.


Assuntos
Anticorpos Antivirais/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Fragmentos Fab das Imunoglobulinas/administração & dosagem , Vacina Antirrábica/administração & dosagem , Vírus da Raiva/efeitos dos fármacos , Raiva/prevenção & controle , Vacinação/métodos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/virologia , Modelos Animais de Doenças , Feminino , Fluoresceína-5-Isotiocianato/química , Corantes Fluorescentes/química , Humanos , Injeções Intraventriculares , Camundongos , Raiva/imunologia , Raiva/mortalidade , Raiva/virologia , Vírus da Raiva/imunologia , Vírus da Raiva/patogenicidade , Análise de Sobrevida , Transfecção/métodos
9.
PLoS One ; 15(5): e0220592, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32469961

RESUMO

Infectious diseases are often transmitted through local interactions. Yet, both surveillance and control measures are implemented within administrative units. Capturing local transmission processes and spatial coupling between regions from aggregate level data is therefore a technical challenge that can shed light on both theoretical questions and practical decisions. Fox rabies has been eliminated from much of Europe through oral rabies vaccination (ORV) programmes. The European Union (EU) co-finances ORV to maintain rabies freedom in EU member and border states via a cordon sanitaire. Models to capture local transmission dynamics and spatial coupling have immediate application to the planning of these ORV campaigns and to other parts of the world considering oral vaccination. We fitted a hierarchical Bayesian state-space model to data on three decades of fox rabies cases and ORV campaigns from Eastern Germany. Specifically, we find that (i) combining regional spatial coupling and heterogeneous local transmission allows us to capture regional rabies dynamics; (ii) incursions from other regions account for less than 1% of cases, but allow for re-emergence of disease; (iii) herd immunity achieved through bi-annual vaccination campaigns is short-lived due to population turnover. Together, these findings highlight the need for regular and sustained vaccination efforts and our modelling approach can be used to provide strategic guidance for ORV delivery. Moreover, we show that biological understanding can be gained from inference from partially observed data on wildlife disease.


Assuntos
Raposas/virologia , Programas de Imunização , Raiva/prevenção & controle , Animais , Animais Selvagens/virologia , Teorema de Bayes , Europa (Continente)/epidemiologia , União Europeia , Alemanha/epidemiologia , Humanos , Raiva/transmissão , Raiva/virologia , Vacina Antirrábica/farmacologia , Vírus da Raiva/efeitos dos fármacos , Vírus da Raiva/patogenicidade
10.
Viruses ; 12(2)2020 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-32033253

RESUMO

Currently, no rabies virus-specific antiviral drugs are available. Ranpirnase has strong antitumor and antiviral properties associated with its ribonuclease activity. TMR-001, a proprietary bulk drug substance solution of ranpirnase, was evaluated against rabies virus in three cell types: mouse neuroblastoma, BSR (baby hamster kidney cells), and bat primary fibroblast cells. When TMR-001 was added to cell monolayers 24 h preinfection, rabies virus release was inhibited for all cell types at three time points postinfection. TMR-001 treatment simultaneous with infection and 24 h postinfection effectively inhibited rabies virus release in the supernatant and cell-to-cell spread with 50% inhibitory concentrations of 0.2-2 nM and 20-600 nM, respectively. TMR-001 was administered at 0.1 mg/kg via intraperitoneal, intramuscular, or intravenous routes to Syrian hamsters beginning 24 h before a lethal rabies virus challenge and continuing once per day for up to 10 days. TMR-001 at this dose, formulation, and route of delivery did not prevent rabies virus transit from the periphery to the central nervous system in this model (n = 32). Further aspects of local controlled delivery of other active formulations or dose concentrations of TMR-001 or ribonuclease analogues should be investigated for this class of drugs as a rabies antiviral therapeutic.


Assuntos
Antivirais/farmacologia , Vírus da Raiva/efeitos dos fármacos , Ribonucleases/farmacologia , Liberação de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Células Cultivadas , Quirópteros , Cricetinae , Feminino , Fibroblastos/virologia , Mesocricetus , Camundongos , Raiva/prevenção & controle , Vírus da Raiva/fisiologia , Ribonucleases/administração & dosagem
11.
Antiviral Res ; 172: 104641, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31672666

RESUMO

Rabies virus (RABV) is a highly neurotropic virus and the causative agent of rabies, an encephalitis with an almost 100% case-fatality rate that remains incurable after the onset of symptoms. Favipiravir (T-705), a broad-spectrum antiviral drug against RNA viruses, has been shown to be effective against RABV in vitro but ineffective in vivo. We hypothesized that favipiravir is effective in infected mice when RABV replicates in the peripheral tissues/nerves but not after virus neuroinvasion. We attempted to clarify this point in this study using in vivo bioluminescence imaging. We generated a recombinant RABV from the field isolate 1088, which expressed red firefly luciferase (1088/RFLuc). This allowed semiquantitative detection and monitoring of primary replication at the inoculation site and viral spread in the central nervous system (CNS) in the same mice. Bioluminescence imaging revealed that favipiravir (300 mg/kg/day) treatment commencing 1 h after intramuscular inoculation of RABV efficiently suppressed viral replication at the inoculation site and the subsequent replication in the CNS. However, virus replication in the CNS was not inhibited when the treatment began 2 days after inoculation. We also found that higher doses (600 or 900 mg/kg/day) of favipiravir could suppress viral replication in the CNS even when administration started 2 days after inoculation. These results support our hypothesis and suggest that a highly effective drug-delivery system into the CNS and/or the enhancement of favipiravir conversion to its active form are required to improve favipiravir treatment of rabies. Furthermore, the bioluminescence imaging system established in this study will facilitate the development of treatment for symptomatic rabies.


Assuntos
Amidas , Sistema Nervoso Central/virologia , Pirazinas , Vírus da Raiva/efeitos dos fármacos , Raiva/tratamento farmacológico , Amidas/administração & dosagem , Amidas/farmacologia , Animais , Antivirais/administração & dosagem , Antivirais/farmacologia , Linhagem Celular , Diagnóstico por Imagem/métodos , Medições Luminescentes , Camundongos , Pirazinas/administração & dosagem , Pirazinas/farmacologia , Carga Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
12.
Virology ; 536: 32-38, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31394410

RESUMO

Rabies continues to poses serious threats to the public health in many countries. The development of novel inexpensive, safe and effective vaccines has become a high priority for rabies control worldwide. We previously generated a novel recombinant rabies vaccine by cloning rabies virus glycoprotein into a chimpanzee adenoviral vector, termed ChAd68-Gp. The present study evaluated the immune responses and protection afforded by this vaccine in beagle dogs. The results demonstrated that intramuscular immunization with both low-dose and high-dose of ChAd68-Gp induced strong immune responses and provided complete protection in beagles even at low-dose. However, when administered orally, high-dose vaccination was protective while low-dose vaccination was ineffective. Further investigation indicated that the low-pH value of gastric juice in the stomach of beagles might decompose the adenovirus. Therefore, suitable formulation for adenovirus-based oral vaccine should be considered and developed. The chimpanzee adenovirus-vectored rabies vaccine ChAd68-Gp warrants extensive test for clinical application.


Assuntos
Adenovirus dos Símios/genética , Anticorpos Neutralizantes/biossíntese , Anticorpos Antivirais/biossíntese , Vacina Antirrábica/administração & dosagem , Vírus da Raiva/efeitos dos fármacos , Raiva/prevenção & controle , Proteínas do Envelope Viral/administração & dosagem , Adenovirus dos Símios/imunologia , Administração Oral , Animais , Cães , Feminino , Vetores Genéticos/química , Vetores Genéticos/imunologia , Concentração de Íons de Hidrogênio , Soros Imunes/administração & dosagem , Injeções Intramusculares , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Raiva/mortalidade , Raiva/patologia , Raiva/virologia , Vacina Antirrábica/genética , Vacina Antirrábica/imunologia , Vírus da Raiva/genética , Vírus da Raiva/imunologia , Vírus da Raiva/patogenicidade , Análise de Sobrevida , Vacinação/métodos , Vacinas Sintéticas , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
13.
J Gen Virol ; 100(8): 1171-1186, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31237530

RESUMO

Rabies virus causes an invariably fatal encephalitis following the onset of clinical disease. Despite the availability of safe and effective vaccines, the clinical stages of rabies encephalitis remain untreatable, with few survivors being documented. A principal obstacle to the treatment of rabies is the neurotropic nature of the virus, with the blood-brain barrier size exclusion limit rendering the delivery of antiviral drugs and molecules to the central nervous system inherently problematic. This review focuses on efforts to try and overcome barriers to molecule delivery to treat clinical rabies and overviews current progress in the development of experimental live rabies virus vaccines that may have future applications in the treatment of clinical rabies, including the attenuation of rabies virus vectors through either the duplication or mutation of existing genes or the incorporation of non-viral elements within the genome. Rabies post-infection treatment (PIT) remains the holy grail of rabies research.


Assuntos
Antivirais/administração & dosagem , Infecções do Sistema Nervoso Central/tratamento farmacológico , Vírus da Raiva/efeitos dos fármacos , Raiva/tratamento farmacológico , Animais , Infecções do Sistema Nervoso Central/virologia , Humanos , Raiva/virologia , Vírus da Raiva/genética , Vírus da Raiva/fisiologia
14.
J Virol ; 93(18)2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31243136

RESUMO

Our previous study showed that pentagalloylglucose (PGG), a naturally occurring hydrolyzable phenolic tannin, possesses significant anti-rabies virus (RABV) activity. In BHK-21 cells, RABV induced the overactivation of signal transducer and activator of transcription 3 (STAT3) by suppressing the expression of suppressor of cytokine signaling 3 (SOCS3). Inhibition of STAT3 by niclosamide, small interfering RNA, or exogenous expression of SOCS3 all significantly suppressed the replication of RABV. Additionally, RABV-induced upregulation of microRNA 455-5p (miR-455-5p) downregulated SOCS3 by directly binding to the 3' untranslated region (UTR) of SOCS3. Importantly, PGG effectively reversed the expression of miR-455-5p and its following SOCS3/STAT3 signaling pathway. Finally, activated STAT3 elicited the expression of interleukin-6 (IL-6), thereby contributing to RABV-associated encephalomyelitis; however, PGG restored the level of IL-6 in vitro and in vivo in a SOCS3/STAT3-dependent manner. Altogether, these data identify a new miR-455-5p/SOCS3/STAT3 signaling pathway that contributes to viral replication and IL-6 production in RABV-infected cells, with PGG exerting its antiviral effect by inhibiting the production of miR-455-5p and the activation of STAT3.IMPORTANCE Rabies virus causes lethal encephalitis in mammals and poses a serious public health threat in many parts of the world. Numerous strategies have been explored to combat rabies; however, their efficacy has always been unsatisfactory. We previously reported a new drug, PGG, which possesses a potent inhibitory activity on RABV replication. Herein, we describe the underlying mechanisms by which PGG exerts its anti-RABV activity. Our results show that RABV induces overactivation of STAT3 in BHK-21 cells, which facilitates viral replication. Importantly, PGG effectively inhibits the activity of STAT3 by disrupting the expression of miR-455-5p and increases the level of SOCS3 by directly targeting the 3' UTR of SOCS3. Furthermore, the downregulated STAT3 inhibits the production of IL-6, thereby contributing to a reduction in the inflammatory response in vivo Our study indicates that PGG effectively inhibits the replication of RABV by the miR-455-5p/SOCS3/STAT3/IL-6-dependent pathway.


Assuntos
Taninos Hidrolisáveis/farmacologia , Vírus da Raiva/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Cricetinae , Interleucina-6/metabolismo , MicroRNAs/efeitos dos fármacos , MicroRNAs/genética , Raiva/virologia , Vírus da Raiva/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo
15.
Virol J ; 16(1): 80, 2019 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-31196105

RESUMO

BACKGROUND: Rabies virus (RABV), a member of Lyssavirus of Rhabdoviridae family, is a kind of negative-strand RNA virus. The zoonosis caused by RABV leads to high mortality in animals and humans. Though with the extensive investigation, the mechanisms of RABV entry into cells have not been well characterized. METHODS: Chemical inhibitors and RNA interference (RNAi) were used to analysis RABV internalization pathway. The expression level of viral N protein was examined by quantitative PCR and western blot, and the virus infection in the cells was visualized by fluorescence microscopy. RESULTS: We firstly examined the endocytosis pathway of the challenge virus standard (CVS) -11 strain in N2a cells. Chlorpromazine treatment and knockdown of clathrin heavy chain (CHC) significantly reduced viral entry, which proved clathrin was required. Meanwhile neither nystatin nor knocking down caveolin-1 (Cav1) in N2a cells had an effect on CVS-11 infection, suggesting that caveolae was independent for CVS-11 internalization. And when cholesterol of cell membrane was extracted by MßCD, viral infection was strongly impacted. Additionally by using the specific inhibitor dynasore and ammonium chloride, we verified that dynamin and a low-pH environment were crucial for RABV infection, which was confirmed by confocal microscopy. CONCLUSIONS: Our results demonstrated that CVS-11 entered N2a cells through a clathrin-mediated, cholesterol-, pH-, dynamin-required, and caveolae-independent endocytic pathway.


Assuntos
Colesterol/metabolismo , Clatrina/metabolismo , Dinaminas/metabolismo , Endocitose , Vírus da Raiva/fisiologia , Internalização do Vírus , Linhagem Celular , Clorpromazina/farmacologia , Concentração de Íons de Hidrogênio , Proteínas do Nucleocapsídeo/genética , Interferência de RNA , Vírus da Raiva/efeitos dos fármacos
16.
Antiviral Res ; 168: 51-60, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31071352

RESUMO

Rabies virus (RABV) is a neurotropic virus that causes fatal encephalitis in humans and animals and still kills up to 59,000 people worldwide every year. To date, only preventive or post-exposure vaccination protects against the disease but therapeutics are missing. After screening a library of 80 kinases inhibitors, we identified two compounds as potent inhibitors of RABV infection: tyrphostin 9 and rottlerin. Mechanism of action studies show that both inhibitors interfere with an early step of viral cycle and can prevent viral replication. In presence of tyrphostin 9, the viral entry through endocytosis is disturbed leading to improper delivery of viral particles in cytoplasm, whereas rottlerin is inhibiting the transcription, most likely by decreasing intracellular ATP concentration, and therefore the replication of the viral genome.


Assuntos
Acetofenonas/farmacologia , Benzopiranos/farmacologia , Nitrilas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Vírus da Raiva/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Animais , Células Cultivadas , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Humanos , RNA Viral/biossíntese , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
17.
Virol Sin ; 34(1): 59-65, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30725320

RESUMO

Exosomes are cell-derived vesicles that are secreted by many eukaryotic cells. It has recently attracted attention as vehicles of intercellular communication. Virus-infected cells release exosomes, which contain viral proteins, RNA, and pathogenic molecules. However, the role of exosomes in virus infection process remains unclear and needs to be further investigated. In this study, we aimed to evaluate the effects of exosomes on rabies virus infection. OptiPrep™ density gradient centrifugation was used to isolate exosomes from rabies virus-infected cell culture supernatants. A rabies virus G protein enzyme-linked immunosorbent assay and acetylcholinesterase activity assays were performed to verify the centrifugation fractions. Exosomes were then characterized using transmission electron microscopy and Western blotting. Our results showed that rabies virus infection increased the release of exosomes. Treatment with GW4869 and si-Rab27a, two exosomal secretion inhibitors, inhibited exosome release. Furthermore, the inhibitors reduced the levels of extracellular and intracellular viral RNA. These data indicated that exosomes may participate in the viral infection process. Moreover, our results establish a basis for future research into the roles of exosomes in rabies virus infection and as potential targets for developing new antiviral strategies.


Assuntos
Exossomos/fisiologia , Interações entre Hospedeiro e Microrganismos , Vírus da Raiva/patogenicidade , Compostos de Anilina/farmacologia , Animais , Compostos de Benzilideno/farmacologia , Centrifugação com Gradiente de Concentração , Chlorocebus aethiops , Meios de Cultura , Células Epiteliais/virologia , Exossomos/ultraestrutura , Microscopia Eletrônica de Transmissão , RNA Viral , Vírus da Raiva/efeitos dos fármacos , Células Vero
18.
Curr Opin Virol ; 35: 1-13, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30753961

RESUMO

Rabies virus (RABV) constitutes a major social and economic burden associated with 60 000 deaths annually worldwide. Although pre-exposure and post-exposure treatment options are available, they are efficacious only when initiated before the onset of clinical symptoms. Aggravating the problem, the current RABV vaccine does not cross-protect against the emerging zoonotic phylogroup II lyssaviruses. A requirement for an uninterrupted cold chain and high cost of the immunoglobulin component of rabies prophylaxis generate an unmet need for the development of RABV-specific antivirals. We discuss desirable anti-RABV drug profiles, past efforts to address the problem and inhibitor candidates identified, and examine how the rapidly expanding structural insight into RABV protein organization has illuminated novel druggable target candidates and paved the way to structure-aided drug optimization. Special emphasis is given to the viral RNA-dependent RNA polymerase complex as a promising target for direct-acting broad-spectrum RABV inhibitors.


Assuntos
Antivirais/farmacologia , Lyssavirus/efeitos dos fármacos , Vírus da Raiva/efeitos dos fármacos , Raiva/tratamento farmacológico , Animais , Anticorpos Antivirais/imunologia , Humanos , Camundongos , Vacinação , Proteínas Virais/genética
19.
J Med Virol ; 91(6): 935-940, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30624794

RESUMO

Neurotropic viruses, such as the rabies virus (RABV) and Japanese encephalitis virus (JEV), induce neuronal dysfunction and complication, causing neuronal damage. Currently, there are still no effective clinical treatments for neuronal injury caused by neurotropic viruses. Memantine, a drug capable of passing through the blood-brain barrier, noncompetitively and reversibly binds to n-methyl- d-aspartic acid (NMDA) receptors. Memantine is used to treat Alzheimer's disease by blocking the activation of extra axonal ion channels, thus preventing neuronal degeneration by inhibiting the abnormal cytosolic Ca 2+ increase. To explore whether memantine can alleviate neurological disturbances caused by RABV and JEV, the following experiments were carried out: (1) for primary neurons cultured in vitro infected with RABV, the addition of memantine showed neuroprotection. (2) In the RABV challenge experiments, memantine had limited therapeutic effect, mildly extending the survival time of mice. In contrast, memantine significantly prolonged the survival time of mice infected with JEV, by reducing the intravascular cuff and inflammatory cell infiltration in mice. Furthermore, memantine decreases the amount of JEV virus in mice brain.


Assuntos
Vírus da Encefalite Japonesa (Espécie)/efeitos dos fármacos , Memantina/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/virologia , Fármacos Neuroprotetores/farmacologia , Vírus da Raiva/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/virologia , Células Cultivadas , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/patologia
20.
Vaccine ; 37(33): 4701-4709, 2019 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-29650384

RESUMO

Rabies is a fatal disease of all mammals causing almost 60,000 human deaths every year. To date, there is no effective treatment of clinical rabies once the symptoms appear. Here, we describe the promising effect of combination therapy composed of molecules that target replication of the rabies virus (RV) at different stages of life cycle and molecules that inhibit some pathways of the innate host immune response accompanied by a blood-brain barrier opener on the outcome of RV infection. The study reports statistically significant extension of survival of mice treated with the drug cocktail containing T-705, ribavirin, interferon α/ß, caspase-1 inhibitor, TNF-α inhibitor, MAPKs inhibitor and HRIG compared to the survival of mice in the virus control group (p = 0.0312). Furthermore, the study points to the significant impact of interferon α/ß on the survival of RV-infected mice. We have shown a significant down regulation of pro-inflammatory molecules (caspase-1 and TNF-a) in the CNS in RV-infected mice treated with a combination of drugs including interferon α/ß.


Assuntos
Antivirais/uso terapêutico , Vírus da Raiva/efeitos dos fármacos , Vírus da Raiva/patogenicidade , Amidas/uso terapêutico , Animais , Anticorpos Antivirais , Modelos Animais de Doenças , Quimioterapia Combinada/métodos , Feminino , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Pirazinas/uso terapêutico , Raiva , Ribavirina/uso terapêutico , Serpinas/uso terapêutico , Proteínas Virais/uso terapêutico , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...