Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 136
Filtrar
1.
Front Cell Infect Microbiol ; 14: 1394713, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38836054

RESUMO

The rabies virus enters the nervous system by interacting with several molecular targets on host cells to modify behavior and trigger receptor-mediated endocytosis of the virion by poorly understood mechanisms. The rabies virus glycoprotein (RVG) interacts with the muscle acetylcholine receptor and the neuronal α4ß2 subtype of the nicotinic acetylcholine receptor (nAChR) family by the putative neurotoxin-like motif. Given that the neurotoxin-like motif is highly homologous to the α7 nAChR subtype selective snake toxin α-bungarotoxin (αBTX), other nAChR subtypes are likely involved. The purpose of this study is to determine the activity of the RVG neurotoxin-like motif on nAChR subtypes that are expressed in brain regions involved in rabid animal behavior. nAChRs were expressed in Xenopus laevis oocytes, and two-electrode voltage clamp electrophysiology was used to collect concentration-response data to measure the functional effects. The RVG peptide preferentially and completely inhibits α7 nAChR ACh-induced currents by a competitive antagonist mechanism. Tested heteromeric nAChRs are also inhibited, but to a lesser extent than the α7 subtype. Residues of the RVG peptide with high sequence homology to αBTX and other neurotoxins were substituted with alanine. Altered RVG neurotoxin-like peptides showed that residues phenylalanine 192, arginine 196, and arginine 199 are important determinants of RVG peptide apparent potency on α7 nAChRs, while serine 195 is not. The evaluation of the rabies ectodomain reaffirmed the observations made with the RVG peptide, illustrating a significant inhibitory impact on α7 nAChR with potency in the nanomolar range. In a mammalian cell culture model of neurons, we confirm that the RVG peptide binds preferentially to cells expressing the α7 nAChR. Defining the activity of the RVG peptide on nAChRs expands our understanding of basic mechanisms in host-pathogen interactions that result in neurological disorders.


Assuntos
Glicoproteínas , Vírus da Raiva , Xenopus laevis , Receptor Nicotínico de Acetilcolina alfa7 , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Animais , Vírus da Raiva/fisiologia , Vírus da Raiva/metabolismo , Humanos , Glicoproteínas/metabolismo , Glicoproteínas/genética , Oócitos/metabolismo , Proteínas Virais/metabolismo , Proteínas Virais/genética , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/genética , Interações Hospedeiro-Patógeno , Ligação Proteica , Raiva/metabolismo , Raiva/virologia , Acetilcolina/metabolismo , Acetilcolina/farmacologia , Neurotoxinas/metabolismo , Neurotoxinas/farmacologia
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.
PLoS One ; 18(11): e0292833, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37922253

RESUMO

The innate immune response is a first-line defense mechanism triggered by rabies virus (RABV). Interferon (IFN) signaling and ISG products have been shown to confer resistance to RABV at various stages of the virus's life cycle. Human tetherin, also known as bone marrow stromal cell antigen 2 (hBST2), is a multifunctional transmembrane glycoprotein induced by IFN that has been shown to effectively counteract many viruses through diverse mechanisms. Here, we demonstrate that hBST2 inhibits RABV budding by tethering new virions to the cell surface. It was observed that release of virus-like particles (VLPs) formed by RABV G (RABV-G VLPs), but not RABV M (RABV-G VLPs), were suppressed by hBST2, indicating that RABV-G has a specific effect on the hBST2-mediated restriction of RABV. The ability of hBST2 to prevent the release of RABV-G VLPs and impede RABV growth kinetics is retained even when hBST2 has mutations at dimerization and/or glycosylation sites, making hBST2 an antagonist to RABV, with multiple mechanisms possibly contributing to the hBST2-mediated suppression of RABV. Our findings expand the knowledge of host antiviral mechanisms that control RABV infection.


Assuntos
Vírus da Raiva , Raiva , Humanos , Vírus da Raiva/fisiologia , Raiva/prevenção & controle , Glicosilação , Asparagina/metabolismo , Cisteína/metabolismo , Dimerização , Liberação de Vírus , Antígeno 2 do Estroma da Médula Óssea/genética , Antígenos CD/metabolismo , Proteínas Ligadas por GPI/metabolismo
4.
Int J Mol Sci ; 24(6)2023 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-36982925

RESUMO

Neurotropic viruses severely damage the central nervous system (CNS) and human health. Common neurotropic viruses include rabies virus (RABV), Zika virus, and poliovirus. When treating neurotropic virus infection, obstruction of the blood-brain barrier (BBB) reduces the efficiency of drug delivery to the CNS. An efficient intracerebral delivery system can significantly increase intracerebral delivery efficiency and facilitate antiviral therapy. In this study, a rabies virus glycopeptide (RVG) functionalized mesoporous silica nanoparticle (MSN) packaging favipiravir (T-705) was developed to generate T-705@MSN-RVG. It was further evaluated for drug delivery and antiviral treatment in a VSV-infected mouse model. The RVG, a polypeptide consisting of 29 amino acids, was conjugated on the nanoparticle to enhance CNS delivery. The T-705@MSN-RVG caused a significant decrease in virus titers and virus proliferation without inducing substantial cell damage in vitro. By releasing T-705, the nanoparticle promoted viral inhibition in the brain during infection. At 21 days post-infection (dpi), a significantly enhanced survival ratio (77%) was observed in the group inoculated with nanoparticle compared with the non-treated group (23%). The viral RNA levels were also decreased in the therapy group at 4 and 6 dpi compared with that of the control group. The T-705@MSN-RVG could be considered a promising system for CNS delivery for treating neurotropic virus infection.


Assuntos
Nanopartículas , Vírus da Raiva , Viroses , Infecção por Zika virus , Zika virus , Humanos , Animais , Camundongos , Vírus da Raiva/fisiologia , Glicopeptídeos , Peptídeos/farmacologia , Antivirais/farmacologia , Antivirais/uso terapêutico
5.
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
6.
Viruses ; 13(8)2021 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-34452292

RESUMO

Rabies virus is a highly neurophilic negative-strand RNA virus with high lethality and remains a huge public health problem in developing countries to date. The double-stranded RNA-binding protein Staufen1 (STAU1) has multiple functions in RNA virus replication, transcription, and translation. However, its function in RABV infection and its mechanism of action are not clear. In this study, we investigated the role of host factor STAU1 in RABV infection of SH-SY-5Y cells. Immunofluorescence, TCID50 titers, confocal microscopy, quantitative real-time PCR and Western blotting were carried out to determine the molecular function and subcellular distribution of STAU1 in these cell lines. Expression of STAU1 in SH-SY-5Y cells was down-regulated by RNA interference or up-regulated by transfection of eukaryotic expression vectors. The results showed that N proficiently colocalized with STAU1 in SH-SY-5Y at 36 h post-infection, and the expression level of STAU1 was also proportional to the time of infection. Down-regulation of STAU1 expression increased the number of Negri body-like structures, enhanced viral replication, and a caused 10-fold increase in viral titers. Meanwhile, N protein and G protein mRNA levels also accumulated gradually with increasing infection time, which implied that STAU1 inhibited rabies virus infection of SH-SY-5Y cells in vitro. In conclusion, our results provide important clues for the detailed replication mechanism of rabies virus and the discovery of therapeutic targets.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Proteínas de Ligação a RNA/metabolismo , Vírus da Raiva/fisiologia , Replicação Viral , Linhagem Celular Tumoral , Proteínas do Citoesqueleto/genética , Interações Hospedeiro-Patógeno , Humanos , Corpos de Inclusão Viral/metabolismo , Proteínas do Nucleocapsídeo/genética , Proteínas do Nucleocapsídeo/metabolismo , Interferência de RNA , Proteínas de Ligação a RNA/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
7.
Front Immunol ; 12: 622516, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33679766

RESUMO

Rabies virus (RABV) is able to reach the central nervous system (CNS) without triggering a strong immune response, using multiple mechanisms to evade and suppress the host immune system. After infection via a bite or scratch from a rabid animal, RABV comes into contact with macrophages, which are the first antigen-presenting cells (APCs) that are recruited to the area and play an essential role in the onset of a specific immune response. It is poorly understood how RABV affects macrophages, and if the interaction contributes to the observed immune suppression. This study was undertaken to characterize the interactions between RABV and human monocyte-derived macrophages (MDMs). We showed that street RABV does not replicate in human MDMs. Using a recombinant trimeric RABV glycoprotein (rRABV-tG) we showed binding to the nicotinic acetylcholine receptor alpha 7 (nAChr α7) on MDMs, and confirmed the specificity using the nAChr α7 antagonist alpha-bungarotoxin (α-BTX). We found that this binding induced the cholinergic anti-inflammatory pathway (CAP), characterized by a significant decrease in tumor necrosis factor α (TNF-α) upon LPS challenge. Using confocal microscopy we found that induction of the CAP is associated with significant cytoplasmic retention of nuclear factor κB (NF-κB). Co-cultures of human MDMs exposed to street RABV and autologous T cells further revealed that the observed suppression of MDMs might affect their function as T cell activators as well, as we found a significant decrease in proliferation of CD8+ T cells and an increased production of the anti-inflammatory cytokine IL-10. Lastly, using flow cytometric analysis we observed a significant increase in expression of the M2-c surface marker CD163, hinting that street RABV might be able to affect macrophage polarization. Taken together, these results show that street RABV is capable of inducing an anti-inflammatory state in human macrophages, possibly affecting T cell functioning.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Macrófagos/imunologia , Vírus da Raiva/fisiologia , Raiva/imunologia , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Anti-Inflamatórios , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Diferenciação Celular , Células Cultivadas , Colinérgicos , Técnicas de Cocultura , Humanos , Interleucina-10/metabolismo , Ativação Linfocitária , NF-kappa B/metabolismo , Neuroimunomodulação , Ligação Proteica , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Células Th2/imunologia
8.
Rev. cuba. med. trop ; 72(1): e457, ene.-abr. 2020. tab, graf
Artigo em Espanhol | LILACS, CUMED | ID: biblio-1126699

RESUMO

Introducción: Las mordeduras de perro afectan fundamentalmente a pacientes en la edad pediátrica, estas engendran peligros como secuelas estéticas y la posibilidad implícita de transmisibilidad del virus de la rabia a seres humanos. Objetivo: caracterizar el comportamiento clínico-epidemiológico de las mordeduras de perro en niños. Métodos: Se realizó un estudio observacional, longitudinal y prospectivo en pacientes con diagnóstico de mordedura de perro con necesidad de inmunización pasiva y activa, ingresados en el Servicio de Miscelánea del Hospital Infantil Sur Antonio María Béguez César de Santiago de Cuba, del 1ro de enero de 2017 al 31 de diciembre de 2018. Resultados: En los pacientes incluidos en el estudio predominaron los del sexo masculino (57,7 por ciento), los grupos de edades de 1 a 5 años 11 meses y 29 días, y de 6 a 10 años 11 meses y 29 días fueron los de mayor incidencia. Aunque no existió asociación estadística entre la localización anatómica de las lesiones y la edad, fueron las de miembros inferiores las más frecuentes en todas las edades con 63 pacientes (40,4 por ciento); destacándose como complicaciones la celulitis, las lesiones óseas y las secuelas estéticas; ningún niño presentó infección rábica. En el 50 por ciento de los casos la mordedura ocurrió en el horario nocturno y los perros de otro domicilio causaron la mayor parte de las agresiones. Conclusiones: El diagnóstico de mordeduras de perro se realizó con mayor frecuencia en niños del sexo masculino de uno y más años, en el horario nocturno y la principal complicación fue la celulitis(AU)


Introduction: Dog bites mainly affect patients in pediatric ages. They pose dangers such as esthetic sequels and the implicit possibility of transmission of the rabies virus to human beings. Objective: Characterize the clinical-epidemiological behavior of dog bites in children. Methods: An observational longitudinal prospective study was conducted of patients diagnosed with dog bite requiring passive and active immunization admitted to the Miscellany Service of Antonio María Béguez César South Children's Hospital in Santiago de Cuba from 1 January 2017 to 31 December 2018. Results: Among the patients included in the study there was a predominance of the male sex (57.7 percent). The age groups 1 to 5 years 11 months and 29 days and 6 to 10 years 11 months and 29 days showed the greatest incidence. Though no statistical association was found between anatomical location of the injuries and age, bites on the lower limbs were the most common at all ages with 63 patients (40.4 percent). Outstanding complications were cellulitis, bone lesions and esthetic sequels. No child had rabies infection. In 50 percent of the cases the bite occurred in the night hours. Most of the attacks were performed by dogs from other households. Conclusions: Dog bite diagnosis was more common among male children aged one year and over. Most bites occurred in the night hours. The main complication was cellulitis(AU)


Assuntos
Humanos , Masculino , Feminino , Lactente , Pré-Escolar , Criança , Mordeduras e Picadas/complicações , Mordeduras e Picadas/diagnóstico , Cães , Vírus da Raiva/fisiologia , Estudos Prospectivos , Estudos Longitudinais , Estudo Observacional
9.
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
10.
PLoS One ; 14(10): e0223684, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31589656

RESUMO

Middle East respiratory syndrome-coronavirus (MERS-CoV) is an emerging virus that causes severe disease with fatal outcomes; however, there are currently no approved vaccines or specific treatments against MERS-CoV. Here, we developed a novel bivalent vaccine against MERS-CoV and rabies virus (RV) using the replication-incompetent P-gene-deficient RV (RVΔP), which has been previously established as a promising and safe viral vector. MERS-CoV spike glycoprotein comprises S1 and S2 subunits, with the S1 subunit being a primary target of neutralizing antibodies. Recombinant RVΔP, which expresses S1 fused with transmembrane and cytoplasmic domains together with 14 amino acids from the ectodomains of the RV-glycoprotein (RV-G), was developed using a reverse genetics method and named RVΔP-MERS/S1. Following generation of RVΔP-MERS/S1 and RVΔP, our analysis revealed that they shared similar growth properties, with the expression of S1 in RVΔP-MERS/S1-infected cells confirmed by immunofluorescence and western blot, and the immunogenicity and pathogenicity evaluated using mouse infection experiments. We observed no rabies-associated signs or symptoms in mice inoculated with RVΔP-MERS/S1. Moreover, virus-specific neutralizing antibodies against both MERS-CoV and RV were induced in mice inoculated intraperitoneally with RVΔP-MERS/S1. These findings indicate that RVΔP-MERS/S1 is a promising and safe bivalent-vaccine candidate against both MERS-CoV and RV.


Assuntos
Infecções por Coronavirus/prevenção & controle , Imunogenicidade da Vacina , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Vírus da Raiva/genética , Vacinas Sintéticas/imunologia , Vacinas Virais/imunologia , Replicação Viral , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Vírus da Raiva/fisiologia , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinas Sintéticas/genética , Células Vero , Vacinas Virais/genética
11.
Adv Virus Res ; 104: 147-183, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31439148

RESUMO

Rhabdoviruses are enveloped viruses with a negative-sense single strand RNA genome and are widespread among a great variety of organisms. In their membrane, they have a single glycoprotein (G) that mediates both virus attachment to cellular receptors and fusion between viral and endosomal membranes allowing viral genome release in the cytoplasm. We present structural and cellular aspects of Rhabdovirus entry into their host cell with a focus on vesicular stomatitis virus (VSV) and rabies virus (RABV) for which the early events of the viral cycle have been extensively studied. Recent data have shown that the only VSV receptors are the members of the LDL-R family. This is in contrast with RABV for which multiple receptors belonging to unrelated families have been identified. Despite having different receptors, after attachment, rhabdovirus internalization occurs through clathrin-mediated endocytosis (CME) in an actin-dependent manner. There are still debates about the exact endocytic pathway of VSV in the cell and on RABV transport in the neuronal axon. In any case, fusion is triggered in the endosomal vesicle via a low-pH induced structural rearrangement of G from its pre- to its postfusion conformation. Vesiculovirus G is one of the best characterized fusion glycoproteins as the previously reported crystal structures of the pre- and postfusion states have been recently completed by those of intermediates during the structural transition. Understanding the entry pathway of rhabdoviruses may have strong impact in biotechnologies as, for example, VSV G is used for pseudotyping lentiviruses to promote efficient transduction, and VSV is a promising oncolytic virus.


Assuntos
Interações Hospedeiro-Patógeno , Vírus da Raiva/fisiologia , Vesiculovirus/fisiologia , Ligação Viral , Internalização do Vírus , Endocitose , Glicoproteínas/metabolismo , Receptores Virais/metabolismo , Proteínas do Envelope Viral/metabolismo
12.
Infect Genet Evol ; 67: 88-100, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30391720

RESUMO

Rabies, caused by the rabies virus (RABV), is the oldest known zoonotic infectious disease. Although the molecular mechanisms of RABV pathogenesis have been investigated extensively, the interactions between host and RABV are not clearly understood. It is now known that long non-coding RNAs (lncRNAs) participate in various physiological and pathological processes, but their possible roles in the host response to RABV infection remain to be elucidated. To better understand the pathogenesis of RABV, RNAs from RABV-infected and uninfected human neuroblastoma cells (SK-N-SH) were analyzed using human lncRNA microarrays. We identified 896 lncRNAs and 579 mRNAs that were differentially expressed after infection, indicating a potential role for lncRNAs in the immune response to RABV. Differentially expressed RNAs were examined using Gene Ontology (GO) analysis and were tentatively assigned to biological pathways using the Kyoto Encyclopedia of Genes and Genomes (KEGG). A lncRNA-mRNA-transcription factor co-expression network was constructed to relate lncRNAs to regulatory factors and pathways that may be important in virus-host interactions. The network analysis suggests that E2F4, TAF7 and several lncRNAs function as transcriptional regulators in various signaling pathways. This study is the first global analysis of lncRNA and mRNA co-expression during RABV infection, provides deeper insight into the mechanism of RABV pathogenesis, and reveals promising candidate for future investigation.


Assuntos
Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , RNA Longo não Codificante , Vírus da Raiva/fisiologia , Raiva/genética , Raiva/virologia , Transcriptoma , Linhagem Celular Tumoral , Biologia Computacional/métodos , Ontologia Genética , Humanos , Neuroblastoma , Análise de Sequência com Séries de Oligonucleotídeos , Interferência de RNA , RNA Mensageiro/genética , Transdução de Sinais , Replicação Viral
13.
Antiviral Res ; 161: 1-9, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30367894

RESUMO

Rabies virus transmits from animals to humans and causes encephalitis. Every year more than 15 million people receive a post exposure prophylaxis (PEP) treatment that is highly effective in the prevention of rabies disease. However, when clinical symptoms appear, for example in people who did not receive PEP, rabies is almost invariably fatal. Due to the limited access to PEP in some target populations, mostly in Asia and in Africa, rabies causes at least 59,000 deaths a year. PEP is not effective after the onset of symptoms and attempts to develop a treatment for clinical rabies have been unsuccessful. After screening a library of 385 FDA-approved drugs, we found that pyrimethamine inhibits rabies infection in vitro through the inhibition of adenosine synthesis. In addition, this compound shows a synergistic interaction with ribavirin. Unfortunately, in rabies infected-mice, pyrimethamine showed no efficacy. One possible explanation may be that the antiviral effect is negated by the observed interference of pyrimethamine with the innate immune response.


Assuntos
Antivirais/farmacologia , Pirimetamina/farmacologia , Vírus da Raiva/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Adenosina/metabolismo , Animais , Sinergismo Farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Vírus da Raiva/fisiologia , Ribavirina/farmacologia , Bibliotecas de Moléculas Pequenas
14.
Biomédica (Bogotá) ; 38(2): 209-215, ene.-jun. 2018. graf
Artigo em Espanhol | LILACS | ID: biblio-950939

RESUMO

Resumen Introducción. Es escasa la información sobre los detalles neuroanatómicos del transporte del virus de la rabia en su ascenso por la médula espinal. Objetivos. Identificar la ruta neuroanatómica de diseminación del virus de la rabia en cada uno de los niveles de la médula espinal de ratón, después de ser inoculado por vía intramuscular. Materiales y métodos. Se inocularon ratones en los músculos isquiotibiales, con virus de la rabia. A partir de las 24 horas después de la inoculación, cada ocho horas se sacrificaron cinco animales por perfusión con paraformaldehído, se les extrajo la médula espinal y se hicieron cortes transversales en los niveles lumbosacro, torácico y cervical. Estos se procesaron mediante inmunohistoquímica para detectar antígenos virales. Resultados. Los primeros antígenos de la rabia se observaron como partículas agregadas, en la médula espinal lumbar, a las 24 horas después de la inoculación, dentro del asta ventral ipsilateral a la extremidad inoculada. A las 32 horas después de la inoculación, se hicieron visibles las primeras motoneuronas inmunorreactivas al virus. A las 40 horas después de la inoculación, se revelaron las primeras neuronas inmunorreactivas en la médula torácica, localizadas en la lámina 8 y, a las 48 horas después de la inoculación en la médula cervical, también en la lámina 8. A las 56 horas después de la inoculación, el virus se había diseminado por toda la médula espinal pero los animales aún no revelaban signos de la enfermedad. Conclusión. En el modelo de ratón aquí utilizado, el virus de la rabia ingresó a la médula espinal por las motoneuronas y, probablemente, utilizó la vía propioespinal descendente para su transporte axonal retrógrado hasta el encéfalo.


Abstract Introduction: Information about the neuroanatomical details of the ascendant transport of the rabies virus through the spinal cord is scarce. Objective: To identify the neuroanatomical route of dissemination of the rabies virus at each of the levels of the spinal cord of mice after being inoculated intramuscularly. Materials and methods: Mice were inoculated with the rabies virus in the hamstrings. After 24 hours post-inoculation, every eight hours, five animals were sacrificed by perfusion with paraformaldehyde. Then, the spinal cord was removed, and transverse cuts were made at the lumbosacral, thoracic, and cervical levels. These were processed by immunohistochemistry for the detection of viral antigens. Results: The first antigens of rabies were observed as aggregated particles in the lumbar spinal cord at 24 hours post-inoculation, within the ventral horn in the same side of the inoculated limb. At 32 hours post inoculation the first motoneurons immunoreactive to the virus became visible. At 40 hours post-inoculation the first immunoreactive neurons were revealed in the thoracic level, located on lamina 8 and at 48 hours post-inoculation in the cervical cord, also on lamina 8. At 56 hours post-inoculation the virus had spread throughout the spinal cord, but the animals still did not show signs of the disease. Conclusion: In the mouse model we used, the rabies virus entered the spinal cord through the motoneurons and probably used the descending propriospinal pathway for its retrograde axonal transport to the encephalus.


Assuntos
Animais , Feminino , Camundongos , Vírus da Raiva/fisiologia , Medula Espinal/virologia , Medula Espinal/anatomia & histologia
15.
mSphere ; 3(2)2018 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-29669880

RESUMO

Enveloped viruses require viral fusion proteins to promote fusion of the viral envelope with a target cell membrane. To drive fusion, these proteins undergo large conformational changes that must occur at the right place and at the right time. Understanding the elements which control the stability of the prefusion state and the initiation of conformational changes is key to understanding the function of these important proteins. The construction of mutations in the fusion protein transmembrane domains (TMDs) or the replacement of these domains with lipid anchors has implicated the TMD in the fusion process. However, the structural and molecular details of the role of the TMD in these fusion events remain unclear. Previously, we demonstrated that isolated paramyxovirus fusion protein TMDs associate in a monomer-trimer equilibrium, using sedimentation equilibrium analytical ultracentrifugation. Using a similar approach, the work presented here indicates that trimeric interactions also occur between the fusion protein TMDs of Ebola virus, influenza virus, severe acute respiratory syndrome coronavirus (SARS CoV), and rabies virus. Our results suggest that TM-TM interactions are important in the fusion protein function of diverse viral families.IMPORTANCE Many important human pathogens are enveloped viruses that utilize membrane-bound glycoproteins to mediate viral entry. Factors that contribute to the stability of these glycoproteins have been identified in the ectodomain of several viral fusion proteins, including residues within the soluble ectodomain. Although it is often thought to simply act as an anchor, the transmembrane domain of viral fusion proteins has been implicated in protein stability and function as well. Here, using a biophysical approach, we demonstrated that the fusion protein transmembrane domains of several deadly pathogens-Ebola virus, influenza virus, SARS CoV, and rabies virus-self-associate. This observation across various viral families suggests that transmembrane domain interactions may be broadly relevant and serve as a new target for therapeutic development.


Assuntos
Glicoproteínas/química , Multimerização Proteica , Proteínas do Envelope Viral/química , Proteínas Virais de Fusão/química , Ebolavirus/química , Ebolavirus/fisiologia , Fusão de Membrana , Orthomyxoviridae/química , Orthomyxoviridae/fisiologia , Domínios Proteicos , Estabilidade Proteica , Vírus da Raiva/química , Vírus da Raiva/fisiologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/química , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Internalização do Vírus
16.
Dev Comp Immunol ; 81: 1-7, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29122634

RESUMO

Though the common vampire bat, Desmodus rotundus, is known as the main rabies virus reservoir in Latin America, no tools are available to investigate its antiviral innate immune system. To characterize the IFN-I pathway, we established an immortalized cell line from a D. rotundus fetal lung named FLuDero. Then we molecularly characterized some of the Toll-like receptors (TLR3, 7, 8 and 9), the three RIG-I-like receptor members, as well as IFNα1 and IFNß. Challenging the FLuDero cell line with poly (I:C) resulted in an up-regulation of both IFNα1 and IFNß and the induction of expression of the different pattern recognition receptors characterized. These findings provide evidence of the intact dsRNA recognition machinery and the IFN-I signaling pathway in our cellular model. Herein, we generated a sum of insightful specific molecular and cellular tools that will serve as a useful model to study virus-host interactions of the common vampire bat.


Assuntos
Quirópteros/imunologia , Proteína DEAD-box 58/genética , Pulmão/citologia , Vírus da Raiva/fisiologia , Receptores Toll-Like/genética , Animais , Linhagem Celular Transformada , Quirópteros/genética , Clonagem Molecular , Reservatórios de Doenças , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Interferon-alfa/metabolismo , Interferon beta/metabolismo , Pulmão/imunologia , Poli I-C/imunologia , RNA de Cadeia Dupla/imunologia , Transdução de Sinais
17.
Antiviral Res ; 149: 89-94, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29122670

RESUMO

Rabies is a lethal viral infection that can affect almost all mammals, including humans. To better understand the replication of Rabies lyssavirus, we investigated if the viral load in brains naturally infected with rabies influences viral internalization and viral growth kinetics in neuroblastoma cells, and if the viral load affects mortality in mice after intradermal infection. We noted that high initial viral loads in brains (group II) were unfavourable for increasing viral titers during serial passages in neuroblastoma cells when compared to low initial viral loads in brains (group I). In addition, group I strains showed higher viral growth and enhanced internalization efficiency in neuroblastoma cells than group II strains. However, we observed that the dominant virus subpopulation in group II promoted efficient viral infection in the central nervous system in the new host, providing a selective advantage to the virus. Our data indicate that rabies infection in animal models depends on not only the virus strain but also the amount of virus. This study may serve as a basis for understanding the biologic proprieties of Rabies lyssavirus strains with respect to the effects on viral replication and the impact on pathogenesis, improving virus yields for use in vaccine development.


Assuntos
Vírus da Raiva/fisiologia , Raiva/virologia , Replicação Viral , Animais , Encéfalo/virologia , Linhagem Celular Tumoral , Células Cultivadas , Camundongos , Neuroblastoma , Carga Viral , Internalização do Vírus
18.
Sci Rep ; 7(1): 12818, 2017 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-28993633

RESUMO

Rabies virus induces drastic behaviour modifications in infected hosts. The mechanisms used to achieve these changes in the host are not known. The main finding of this study is that a region in the rabies virus glycoprotein, with homologies to snake toxins, has the ability to alter behaviour in animals through inhibition of nicotinic acetylcholine receptors present in the central nervous system. This finding provides a novel aspect to virus receptor interaction and host manipulation by pathogens in general. The neurotoxin-like region of the rabies virus glycoprotein inhibited acetylcholine responses of α4ß2 nicotinic receptors in vitro, as did full length ectodomain of the rabies virus glycoprotein. The same peptides significantly altered a nicotinic receptor induced behaviour in C. elegans and increased locomotor activity levels when injected into the central nervous system of mice. These results provide a mechanistic explanation for the behavioural changes in hosts infected by rabies virus.


Assuntos
Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/virologia , Glicoproteínas/química , Interações Hospedeiro-Patógeno , Vírus da Raiva/fisiologia , Receptores de Neurotransmissores/antagonistas & inibidores , Venenos de Serpentes/química , Sequência de Aminoácidos , Animais , Comportamento Animal , Caenorhabditis elegans/virologia , Sequência Conservada , Humanos , Camundongos , Neurotoxinas/química , Neurotoxinas/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Ligação Proteica , Domínios Proteicos , Receptores de Neurotransmissores/metabolismo , Receptores Nicotínicos/química , Receptores Nicotínicos/metabolismo , Homologia de Sequência de Aminoácidos , Xenopus
19.
Infect Genet Evol ; 56: 44-53, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29056542

RESUMO

It was reported that ISG15 and its activating enzyme UBA7 have antiviral functions. However, there is no study to demonstrate whether ISG15 and UBA7 have anti-rabies virus function. In the current study, In vivo and in vitro anti-rabies virus function of ISG15 and UBA7 were investigated using RNAi technology. The results showed that shRNA knock-down of expression of ISG15 and UBA7 increased the titers of RABV in neuroblastoma cell line NA and microglial cell line BV-2 cells and shRNA knockdown of ISG15 conjugation alleviates the IFN-induced inhibition of RABV gene expression in vitro. Lentiviral vector mediated-shRNA knock-down of expression of ISG15 and UBA7 increased the titers of RABV in mouse brains and decreased the survivorship of mice. The study showed that ISG15 and UBA7 inhibit RABV replication in vitro and in vivo. To our knowledge, we for the first time documented the anti-RABV function of ISG15 and UBA7, which may provide a means of understanding the pathogenesis of rabies and improving therapeutic methods.


Assuntos
Citocinas/metabolismo , Interações Hospedeiro-Patógeno , Vírus da Raiva/fisiologia , Raiva/metabolismo , Raiva/virologia , Enzimas Ativadoras de Ubiquitina/metabolismo , Replicação Viral , Animais , Linhagem Celular Tumoral , Citocinas/genética , Feminino , Expressão Gênica , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Camundongos , Plasmídeos/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Raiva/genética , Enzimas Ativadoras de Ubiquitina/genética , Ubiquitinas/genética , Ubiquitinas/metabolismo
20.
Braz. j. microbiol ; 48(3): 566-569, July-Sept. 2017. tab, graf
Artigo em Inglês | LILACS | ID: biblio-889146

RESUMO

Abstract The aim of this study was to assess the in vitro and in vivo effects of short-interfering RNAs (siRNAs) against rabies virus phosphoprotein (P) mRNA in a post-infection treatment for rabies as an extension of a previous report (Braz J Microbiol. 2013 Nov 15;44(3):879-82). To this end, rabies virus strain RABV-4005 (related to the Desmodus rotundus vampire bat) were used to inoculate BHK-21 cells and mice, and the transfection with each of the siRNAs was made with Lipofectamine-2000™. In vitro results showed that siRNA 360 was able to inhibit the replication of strain RABV-4005 with a 1 log decrease in virus titter and 5.16-fold reduction in P mRNA, 24 h post-inoculation when compared to non-treated cells. In vivo, siRNA 360 was able to induce partial protection, but with no significant difference when compared to non-treated mice. These results indicate that, despite the need for improvement for in vivo applications, P mRNA might be a target for an RNAi-based treatment for rabies.


Assuntos
Animais , Fosfoproteínas/genética , Raiva/veterinária , Vírus da Raiva/genética , Proteínas Virais/genética , Quirópteros/virologia , RNA Interferente Pequeno/genética , Interferência de RNA , Fosfoproteínas/metabolismo , Raiva/virologia , Vírus da Raiva/fisiologia , Proteínas Virais/metabolismo , Replicação Viral , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA