Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 164
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
J Virol ; 97(3): e0012523, 2023 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-36786631

RESUMO

Tacaribe virus (TCRV) is the prototype of New World mammarenaviruses, a group that includes several members that cause hemorrhagic fevers in humans. The TCRV genome comprises two RNA segments, named S (small) and L (large). Both genomic segments contain noncoding regions (NCRs) at their 5' and 3' ends. While the 5'- and 3'-terminal 19-nucleotide sequences are known to be essential for promoter function, the role of their neighboring internal noncoding region (iNCR) sequences remains poorly understood. To analyze the relevance of the 5' and 3' iNCRs in TCRV S RNA synthesis, mutant S-like minigenomes and miniantigenomes were generated. Using a minireplicon assay, Northern blotting, and reverse transcription-quantitative PCR, we demonstrated that the genomic 5' iNCR is specifically engaged in minigenome replication yet is not directly involved in minigenome transcription, and we showed that the S genome 3' iNCR is barely engaged in this process. Analysis of partial deletions and point mutations, as well as total or partial substitution of the 5' iNCR sequence, led us to conclude that the integrity of the whole genomic 5' iNCR is essential and that a local predicted secondary structure or RNA-RNA interactions between the 5' and 3' iNCRs are not strictly required for viral S RNA synthesis. Furthermore, we employed a TCRV reverse genetic approach to ask whether manipulation of the S genomic 5' iNCR sequence may be suitable for viral attenuation. We found that mutagenesis of the 5' promoter-proximal subregion slightly impacted recombinant TCRV virulence in vivo. IMPORTANCE The Mammarenavirus genus of the Arenaviridae family includes several members that cause severe hemorrhagic fevers associated with high morbidity and mortality rates, for which no FDA-approved vaccines and limited therapeutic resources are available. We provide evidence demonstrating the specific involvement of the TCRV S 5' noncoding sequence adjacent to the viral promoter in replication. In addition, we examined the relevance of this region in the context of an in vivo infection. Our findings provide insight into the mechanism through which this 5' viral RNA noncoding region assists the L polymerase for efficient viral S RNA synthesis. Also, these findings expand our understanding of the effect of genetic manipulation of New World mammarenavirus sequences aimed at the rational design of attenuated recombinant virus vaccine platforms.


Assuntos
Arenavirus do Novo Mundo , Genoma Viral , Replicação do RNA , Humanos , Arenavirus do Novo Mundo/genética , Arenavirus do Novo Mundo/patogenicidade , RNA Viral/genética , Replicação do RNA/genética , Mutagênese , Regiões Promotoras Genéticas/genética
2.
PLoS Pathog ; 17(3): e1009356, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33647064

RESUMO

Several arenaviruses cause hemorrhagic fevers in humans with high case fatality rates. A vaccine named Candid#1 is available only against Junin virus (JUNV) in Argentina. Specific N-linked glycans on the arenavirus surface glycoprotein (GP) mask important epitopes and help the virus evade antibody responses. However the role of GPC glycans in arenavirus pathogenicity is largely unclear. In a lethal animal model of hemorrhagic fever-causing Machupo virus (MACV) infection, we found that a chimeric MACV with the ectodomain of GPC from Candid#1 vaccine was partially attenuated. Interestingly, mutations resulting in acquisition of N-linked glycans at GPC N83 and N166 frequently occurred in late stages of the infection. These glycosylation sites are conserved in the GPC of wild-type MACV, indicating that this is a phenotypic reversion for the chimeric MACV to gain those glycans crucial for infection in vivo. Further studies indicated that the GPC mutant viruses with additional glycans became more resistant to neutralizing antibodies and more virulent in animals. On the other hand, disruption of these glycosylation sites on wild-type MACV GPC rendered the virus substantially attenuated in vivo and also more susceptible to antibody neutralization, while loss of these glycans did not affect virus growth in cultured cells. We also found that MACV lacking specific GPC glycans elicited higher levels of neutralizing antibodies against wild-type MACV. Our findings revealed the critical role of specific glycans on GPC in arenavirus pathogenicity and have important implications for rational design of vaccines against this group of hemorrhagic fever-causing viruses.


Assuntos
Anticorpos Antivirais/imunologia , Arenavirus/imunologia , Febre Hemorrágica Americana/virologia , Vírus Junin/patogenicidade , Animais , Anticorpos Neutralizantes/imunologia , Arenavirus do Novo Mundo/genética , Arenavirus do Novo Mundo/imunologia , Arenavirus do Novo Mundo/patogenicidade , Febre Hemorrágica Americana/imunologia , Febre Hemorrágica Americana/prevenção & controle , Humanos , Vírus Junin/imunologia , Vacinas Virais/imunologia
3.
PLoS Biol ; 17(2): e3000137, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30726215

RESUMO

Tripartite motif (TRIM) proteins belong to a large family with many roles in host biology, including restricting virus infection. Here, we found that TRIM2, which has been implicated in cases of Charcot-Marie-Tooth disease (CMTD) in humans, acts by blocking hemorrhagic fever New World arenavirus (NWA) entry into cells. We show that Trim2-knockout mice, as well as primary fibroblasts from a CMTD patient with mutations in TRIM2, are more highly infected by the NWAs Junín and Tacaribe virus than wild-type mice or cells are. Using mice with different Trim2 gene deletions and TRIM2 mutant constructs, we demonstrate that its antiviral activity is uniquely independent of the RING domain encoding ubiquitin ligase activity. Finally, we show that one member of the TRIM2 interactome, signal regulatory protein α (SIRPA), a known inhibitor of phagocytosis, also restricts NWA infection and conversely that TRIM2 limits phagocytosis of apoptotic cells. In addition to demonstrating a novel antiviral mechanism for TRIM proteins, these studies suggest that the NWA entry and phagocytosis pathways overlap.


Assuntos
Antígenos de Diferenciação/genética , Arenavirus do Novo Mundo/genética , Doença de Charcot-Marie-Tooth/genética , Interações Hospedeiro-Patógeno/genética , Proteínas Nucleares/genética , Receptores Imunológicos/genética , Animais , Antígenos de Diferenciação/imunologia , Antígenos de Diferenciação/metabolismo , Apoptose , Arenavirus do Novo Mundo/crescimento & desenvolvimento , Arenavirus do Novo Mundo/patogenicidade , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/virologia , Linhagem Celular Tumoral , Doença de Charcot-Marie-Tooth/metabolismo , Doença de Charcot-Marie-Tooth/patologia , Chlorocebus aethiops , Fibroblastos/imunologia , Fibroblastos/metabolismo , Fibroblastos/virologia , Regulação da Expressão Gênica , Células HEK293 , Interações Hospedeiro-Patógeno/imunologia , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/imunologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/imunologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas de Neurofilamentos/genética , Proteínas de Neurofilamentos/imunologia , Proteínas de Neurofilamentos/metabolismo , Proteínas Nucleares/imunologia , Proteínas Nucleares/metabolismo , Osteoblastos/imunologia , Osteoblastos/metabolismo , Osteoblastos/virologia , Cultura Primária de Células , Receptores Imunológicos/imunologia , Receptores Imunológicos/metabolismo , Transdução de Sinais , Células Vero , Internalização do Vírus
4.
J Virol ; 91(20)2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28794024

RESUMO

The arenavirus family consists of several highly pathogenic viruses, including the Old World (OW) arenavirus Lassa fever virus (LASV) and the New World (NW) Junin virus (JUNV) and Machupo virus (MACV). Host response to infection by these pathogenic arenaviruses is distinct in many aspects. JUNV and MACV infections readily induce an interferon (IFN) response in human cells, while LASV infection usually triggers an undetectable or weak IFN response. JUNV induces an IFN response through RIG-I, suggesting that the host non-self RNA sensor readily detects JUNV viral RNAs (vRNAs) during infection and activates IFN response. Double-stranded-RNA (dsRNA)-activated protein kinase R (PKR) is another host non-self RNA sensor classically known for its vRNA recognition activity. Here we report that infection with NW arenaviruses JUNV and MACV, but not OW LASV, activated PKR, concomitant with elevated phosphorylation of the translation initiation factor α subunit of eukaryotic initiation factor 2 (eIF2α). Host protein synthesis was substantially suppressed in MACV- and JUNV-infected cells but was only marginally reduced in LASV-infected cells. Despite the antiviral activity known for PKR against many other viruses, the replication of JUNV and MACV was not impaired but was slightly augmented in wild-type (wt) cells compared to that in PKR-deficient cells, suggesting that PKR or PKR activation did not negatively affect JUNV and MACV infection. Additionally, we found an enhanced IFN response in JUNV- or MACV-infected PKR-deficient cells, which was inversely correlated with virus replication.IMPORTANCE The detection of viral RNA by host non-self RNA sensors, including RIG-I and MDA5, is critical to the initiation of the innate immune response to RNA virus infection. Among pathogenic arenaviruses, the OW LASV usually does not elicit an interferon response. However, the NW arenaviruses JUNV and MACV readily trigger an IFN response in a RIG-I-dependent manner. Here, we demonstrate for the first time that pathogenic NW arenaviruses JUNV and MACV, but not the OW arenavirus LASV, activated the dsRNA-dependent PKR, another host non-self RNA sensor, during infection. Interestingly, the replication of JUNV and MACV was not restricted but was rather slightly augmented in the presence of PKR. Our data provide new evidence for a distinct interplay between host non-self RNA sensors and pathogenic arenaviruses, which also provides insights into the pathogenesis of arenaviruses and may facilitate the design of vaccines and treatments against arenavirus-caused diseases.


Assuntos
Arenavirus do Novo Mundo/patogenicidade , Arenavirus do Velho Mundo/patogenicidade , Imunidade Inata , Vírus Junin/patogenicidade , Receptores de Reconhecimento de Padrão/metabolismo , Replicação Viral , eIF-2 Quinase/metabolismo , Células A549 , Arenavirus do Novo Mundo/fisiologia , Arenavirus do Velho Mundo/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Interferons/biossíntese , Interferons/imunologia , Vírus Junin/fisiologia , Fosforilação , Receptores de Reconhecimento de Padrão/genética , Fatores de Transcrição/metabolismo , eIF-2 Quinase/genética
6.
J Virol ; 90(3): 1290-7, 2016 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-26581982

RESUMO

UNLABELLED: Machupo virus (MACV) is the causative agent of Bolivian hemorrhagic fever. Our previous study demonstrated that a MACV strain with a single amino acid substitution (F438I) in the transmembrane domain of glycoprotein is attenuated but genetically unstable in mice. MACV is closely related to Junin virus (JUNV), the causative agent of Argentine hemorrhagic fever. Others and our group have identified the glycoprotein to be the major viral factor determining JUNV attenuation. In this study, we tested the compatibility of the glycoprotein of the Candid#1 live-attenuated vaccine strain of JUNV in MACV replication and its ability to attenuate MACV in vivo. Recombinant MACV with the Candid#1 glycoprotein (rMACV/Cd#1-GPC) exhibited growth properties similar to those of Candid#1 and was genetically stable in vitro. In a mouse model of lethal infection, rMACV/Cd#1-GPC was fully attenuated, more immunogenic than Candid#1, and fully protective against MACV infection. Therefore, the MACV strain expressing the glycoprotein of Candid#1 is safe, genetically stable, and highly protective against MACV infection in a mouse model. IMPORTANCE: Currently, there are no FDA-approved vaccines and/or treatments for Bolivian hemorrhagic fever, which is a fatal human disease caused by MACV. The development of antiviral strategies to combat viral hemorrhagic fevers, including Bolivian hemorrhagic fever, is one of the top priorities of the Implementation Plan of the U.S. Department of Health and Human Services Public Health Emergency Medical Countermeasures Enterprise. Here, we demonstrate for the first time that MACV expressing glycoprotein of Candid#1 is a safe, genetically stable, highly immunogenic, and protective vaccine candidate against Bolivian hemorrhagic fever.


Assuntos
Arenavirus do Novo Mundo/genética , Arenavirus do Novo Mundo/imunologia , Glicoproteínas de Membrana/genética , Recombinação Genética , Proteínas do Envelope Viral/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia , Estruturas Animais/patologia , Animais , Arenavirus do Novo Mundo/patogenicidade , Peso Corporal , Modelos Animais de Doenças , Instabilidade Genômica , Febre Hemorrágica Americana/patologia , Febre Hemorrágica Americana/prevenção & controle , Histocitoquímica , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Análise de Sequência de DNA , Análise de Sobrevida , Temperatura , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Virais/genética , Virulência
7.
PLoS Biol ; 11(5): e1001571, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23723737

RESUMO

Transferrin Receptor (TfR1) is the cell-surface receptor that regulates iron uptake into cells, a process that is fundamental to life. However, TfR1 also facilitates the cellular entry of multiple mammalian viruses. We use evolutionary and functional analyses of TfR1 in the rodent clade, where two families of viruses bind this receptor, to mechanistically dissect how essential housekeeping genes like TFR1 successfully balance the opposing selective pressures exerted by host and virus. We find that while the sequence of rodent TfR1 is generally conserved, a small set of TfR1 residue positions has evolved rapidly over the speciation of rodents. Remarkably, all of these residues correspond to the two virus binding surfaces of TfR1. We show that naturally occurring mutations at these positions block virus entry while simultaneously preserving iron-uptake functionalities, both in rodent and human TfR1. Thus, by constantly replacing the amino acids encoded at just a few residue positions, TFR1 divorces adaptation to ever-changing viruses from preservation of key cellular functions. These dynamics have driven genetic divergence at the TFR1 locus that now enforces species-specific barriers to virus transmission, limiting both the cross-species and zoonotic transmission of these viruses.


Assuntos
Genes Essenciais , Receptores da Transferrina/metabolismo , Sequência de Aminoácidos , Animais , Arenavirus do Novo Mundo/genética , Arenavirus do Novo Mundo/patogenicidade , Linhagem Celular , Cães , Humanos , Ferro/metabolismo , Camundongos , Dados de Sequência Molecular , Mutação , Polimorfismo de Nucleotídeo Único , Receptores da Transferrina/genética , Receptores Virais/química , Receptores Virais/metabolismo , Proteínas Virais/química , Proteínas Virais/metabolismo , Internalização do Vírus , Zoonoses
8.
PLoS Biol ; 11(5): e1001574, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23723739

RESUMO

All viruses need to bind to specific receptor molecules on the surface of target cells to initiate infection. Virus-receptor binding is highly specific, and this specificity determines both the species and the cell type that can be infected by a given virus. In some well-studied cases, the virus-binding region on the receptor has been found to be unrelated to the receptor's normal cellular function. Resistance to virus infection can thus evolve by selection of mutations that alter amino acids in the binding region with minimal effect on normal function. This sort of positive selection can be used to infer the history of the host-virus "arms race" during their coevolution. In a new study, Demogines et al. use a combination of phylogenetic, structural, and virological analysis to infer the history and significance of positive selection on the transferrin receptor TfR1, a housekeeping protein required for iron uptake and the cell surface receptor for at least three different types of virus. The authors show that only two parts of the rodent TfR1 molecule have been subject to positive selection and that these correspond to the binding sites for two of these viruses-the mouse mammary tumor virus (a retrovirus) and Machupo virus (an arenavirus). They confirmed this result by introducing the inferred binding site mutations into the wild-type protein and testing for receptor function. Related arenaviruses are beginning to spread in human populations in South America as the cause of often fatal hemorrhagic fevers, and, although Demogines et al. could find no evidence of TfR1 mutations in this region that might have been selected as a consequence of human infection, the authors identified one such mutation in Asian populations that affects infection with these viruses.


Assuntos
Interações Hospedeiro-Patógeno , Receptores Virais/metabolismo , Vírion/patogenicidade , Animais , Arenavirus do Novo Mundo/metabolismo , Arenavirus do Novo Mundo/patogenicidade , Sítios de Ligação , Humanos , Vírus do Tumor Mamário do Camundongo/genética , Vírus do Tumor Mamário do Camundongo/metabolismo , Camundongos , Filogenia , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , Receptores Virais/genética , Vírion/metabolismo
9.
Vet Pathol ; 53(1): 190-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26139838

RESUMO

Machupo virus, the cause of Bolivian hemorrhagic fever, is a highly lethal viral hemorrhagic fever with no Food and Drug Administration-approved vaccines or therapeutics. This study evaluated the guinea pig as a model using the Machupo virus-Chicava strain administered via aerosol challenge. Guinea pigs (Cavia porcellus) were serially sampled to evaluate the temporal progression of infection, gross and histologic lesions, and sequential changes in serum chemistry and hematology. The incubation period was 5 to 12 days, and complete blood counts revealed leukopenia with lymphopenia and thrombocytopenia. Gross pathologic findings included congestion and hemorrhage of the gastrointestinal mucosa and serosa, noncollapsing lungs with fluid exudation, enlarged lymph nodes, and progressive pallor and friability of the liver. Histologic lesions consisted of foci of degeneration and cell death in the haired skin, liver, pancreas, adrenal glands, lymph nodes, tongue, esophagus, salivary glands, renal pelvis, small intestine, and large intestine. Lymphohistiocytic interstitial pneumonia was also present. Inflammation within the central nervous system, interpreted as nonsuppurative encephalitis, was histologically apparent approximately 16 days postexposure and was generally progressive. Macrophages in the tracheobronchial lymph node, on day 5 postexposure, were the first cells to demonstrate visible viral antigen. Viral antigen was detected throughout the lymphoid system by day 9 postexposure, followed by prominent spread within epithelial tissues and then brain. This study provides insight into the course of Machupo virus infection and supports the utility of guinea pigs as an additional animal model for vaccine and therapeutic development.


Assuntos
Arenavirus do Novo Mundo/patogenicidade , Modelos Animais de Doenças , Cobaias , Febre Hemorrágica Americana/patologia , Glândulas Suprarrenais/patologia , Aerossóis , Animais , Epitélio/patologia , Feminino , Febre Hemorrágica Americana/virologia , Humanos , Fígado/patologia , Pulmão/patologia , Linfonodos/patologia , Masculino , Pâncreas/patologia
10.
J Virol ; 88(18): 10995-9, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25031335

RESUMO

Machupo virus (MACV) is the etiologic agent of Bolivian hemorrhagic fever (BHF). Utilizing a reverse-genetics system recently developed, we report the rescue of a rationally modified recombinant MACV containing a single mutation in the transmembrane region of the glycoprotein. Following challenge of susceptible mice, we identified a significant reduction in virulence in the novel virus. We also identified an instability leading to reversion of the single mutation to a wild-type genotype.


Assuntos
Substituição de Aminoácidos , Arenavirus do Novo Mundo/metabolismo , Arenavirus do Novo Mundo/patogenicidade , Membrana Celular/virologia , Glicoproteínas/genética , Febre Hemorrágica Americana/virologia , Mutação de Sentido Incorreto , Proteínas Virais/química , Proteínas Virais/genética , Sequência de Aminoácidos , Animais , Arenavirus do Novo Mundo/química , Arenavirus do Novo Mundo/genética , Sequência de Bases , Glicoproteínas/química , Glicoproteínas/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Proteínas Virais/metabolismo , Virulência
11.
J Gen Virol ; 95(Pt 1): 1-15, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24068704

RESUMO

Arenaviruses can cause fatal human haemorrhagic fever (HF) diseases for which vaccines and therapies are extremely limited. Both the New World (NW) and Old World (OW) groups of arenaviruses contain HF-causing pathogens. Although these two groups share many similarities, important differences with regard to pathogenicity and molecular mechanisms of virus infection exist. These closely related pathogens share many characteristics, including genome structure, viral assembly, natural host selection and the ability to interfere with innate immune signalling. However, members of the NW and OW viruses appear to use different receptors for cellular entry, as well as different mechanisms of virus internalization. General differences in disease signs and symptoms and pathological lesions in patients infected with either NW or OW arenaviruses are also noted and discussed herein. Whilst both the OW Lassa virus (LASV) and the NW Junin virus (JUNV) can cause disruption of the vascular endothelium, which is an important pathological feature of HF, the immune responses to these related pathogens seem to be quite distinct. Whereas LASV infection results in an overall generalized immune suppression, patients infected with JUNV seem to develop a cytokine storm. Additionally, the type of immune response required for recovery and clearance of the virus is different between NW and OW infections. These differences may be important to allow the viruses to evade host immune detection. Understanding these differences will aid the development of new vaccines and treatment strategies against deadly HF viral infections.


Assuntos
Infecções por Arenaviridae/patologia , Infecções por Arenaviridae/virologia , Arenavirus do Novo Mundo/genética , Arenavirus do Velho Mundo/genética , Febres Hemorrágicas Virais/patologia , Febres Hemorrágicas Virais/virologia , Animais , Infecções por Arenaviridae/imunologia , Arenavirus do Novo Mundo/classificação , Arenavirus do Novo Mundo/imunologia , Arenavirus do Novo Mundo/patogenicidade , Arenavirus do Velho Mundo/classificação , Arenavirus do Velho Mundo/imunologia , Arenavirus do Velho Mundo/patogenicidade , Febres Hemorrágicas Virais/imunologia , Humanos
12.
Artigo em Russo | MEDLINE | ID: mdl-25286529

RESUMO

AIM: Study sensitivity of laboratory animals to a causative agent ofArgentine hemorrhagic fever. MATERIALS AND METHODS: Junin virus strain XJ P37 was obtained from the State Collection of Causative Agents of Viral Hemorrhagic Fevers of the Pathogenicity Group I of Scientific Research Center of the 33rd Central Scientific Research Test Institute (SRC of the 33rd CSRTI). Junin virus strain XJ P37 culture with biological activity of 5.2 1g PFU x ml was used in the experiments. Mice (2 - 4 and 7 - 14 days old), guinea pigs (250 - 300 g), 1.8 - 2.5 kg shinshilla breed rabbits, 2.0 - 3.0 kg javanese macaque monkeys were obtained from vivarium of the SRC of the 33rd CSRTI. Vero (B) and GMK-AH-1 (D) cell cultures were obtained from cell culture collection of the SRC of the 33rd CSRTI. Biological activity calculation of Junin virus was carried out by Kerber in I.P. Amsharin modification. RESULTS: Lethality in animals was from 12.5 to 50% after intranasal and intraperitoneal infection of guinea pigs, intramuscular, intraperitoneal and subcutaneous infection of rabbits, intracerebral and intranasal infection of mice at the doses from 0.4 to 1.0 x 10(5) PFU. Death of infected monkeys after intramuscular administration of the virus at 1.0 x 10(4) PFU dose was not observed. In 100% of surviving animals formation of virus-neutralizing antibodies was registered. CONCLUSION: Evaluation of sensitivity of laboratory animals to Junin virus has shown that intracerebrally infected mice may be used to maintain causative agent culture, infected guinea pigs - to prepare virus-containing cultures and modelling infection exacerbation in humans. Intramuscularly infected rabbits may be used to obtain hyper-immune sera.


Assuntos
Arenavirus do Novo Mundo/patogenicidade , Febre Hemorrágica Americana/virologia , Vírus Junin/patogenicidade , Animais , Anticorpos Antivirais/isolamento & purificação , Modelos Animais de Doenças , Cobaias , Febre Hemorrágica Americana/epidemiologia , Febre Hemorrágica Americana/patologia , Humanos , Camundongos , Coelhos
13.
J Virol ; 86(20): 11301-10, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22875974

RESUMO

Arenaviruses are responsible for acute hemorrhagic fevers with high mortality and pose significant threats to public health and biodefense. These enveloped negative-sense RNA viruses replicate in the cell cytoplasm and express four proteins. To better understand how these proteins insinuate themselves into cellular processes to orchestrate productive viral replication, we have identified and characterized novel cytosolic structures involved in arenavirus replication and transcription. In cells infected with the nonpathogenic Tacaribe virus or the attenuated Candid#1 strain of Junín virus, we find that newly synthesized viral RNAs localize to cytosolic puncta containing the nucleoprotein (N) of the virus. Density gradient centrifugation studies reveal that these replication-transcription complexes (RTCs) are associated with cellular membranes and contain full-length genomic- and antigenomic-sense RNAs. Viral mRNAs segregate at a higher buoyant density and are likewise scant in immunopurified RTCs, consistent with their translation on bulk cellular ribosomes. In addition, confocal microscopy analysis reveals that RTCs contain the lipid phosphatidylinositol-4-phosphate and proteins involved in cellular mRNA metabolism, including the large and small ribosomal subunit proteins L10a and S6, the stress granule protein G3BP1, and a subset of translation initiation factors. Elucidating the structure and function of RTCs will enhance our understanding of virus-cell interactions that promote arenavirus replication and mitigate against host cell immunity. This knowledge may lead to novel intervention strategies to limit viral virulence and pathogenesis.


Assuntos
Infecções por Arenaviridae/virologia , Arenavirus do Novo Mundo/fisiologia , Citosol/virologia , Vírus Junin/fisiologia , RNA Viral/biossíntese , Replicação Viral , Animais , Arenavirus do Novo Mundo/patogenicidade , Membrana Celular/virologia , Centrifugação com Gradiente de Concentração , Chlorocebus aethiops , Citosol/ultraestrutura , Vírus Junin/patogenicidade , Nucleoproteínas , Fosfatos de Fosfatidilinositol/análise , RNA Viral/genética , Transcrição Gênica , Células Vero
14.
J Virol ; 86(15): 8185-97, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22623788

RESUMO

Arenaviruses include several causative agents of hemorrhagic fever (HF) disease in humans that are associated with high morbidity and significant mortality. Morbidity and lethality associated with HF arenaviruses are believed to involve the dysregulation of the host innate immune and inflammatory responses that leads to impaired development of protective and efficient immunity. The molecular mechanisms underlying this dysregulation are not completely understood, but it is suggested that viral infection leads to disruption of early host defenses and contributes to arenavirus pathogenesis in humans. We demonstrate in the accompanying paper that the prototype member in the family, lymphocytic choriomeningitis virus (LCMV), disables the host innate defense by interfering with type I interferon (IFN-I) production through inhibition of the interferon regulatory factor 3 (IRF3) activation pathway and that the viral nucleoprotein (NP) alone is responsible for this inhibitory effect (C. Pythoud, W. W. Rodrigo, G. Pasqual, S. Rothenberger, L. Martínez-Sobrido, J. C. de la Torre, and S. Kunz, J. Virol. 86:7728-7738, 2012). In this report, we show that LCMV-NP, as well as NPs encoded by representative members of both Old World (OW) and New World (NW) arenaviruses, also inhibits the nuclear translocation and transcriptional activity of the nuclear factor kappa B (NF-κB). Similar to the situation previously reported for IRF3, Tacaribe virus NP (TCRV-NP) does not inhibit NF-κB nuclear translocation and transcriptional activity to levels comparable to those seen with other members in the family. Altogether, our findings demonstrate that arenavirus infection inhibits NF-κB-dependent innate immune and inflammatory responses, possibly playing a key role in the pathogenesis and virulence of arenavirus.


Assuntos
Arenavirus do Novo Mundo/imunologia , Imunidade Inata , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , NF-kappa B/imunologia , Nucleoproteínas/imunologia , Proteínas Virais/imunologia , Animais , Arenavirus do Novo Mundo/genética , Arenavirus do Novo Mundo/patogenicidade , Chlorocebus aethiops , Cricetinae , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/patologia , Coriomeningite Linfocítica/transmissão , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/patogenicidade , NF-kappa B/genética , Nucleoproteínas/genética , Células Vero , Proteínas Virais/genética
15.
Virol J ; 10: 221, 2013 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-23816343

RESUMO

BACKGROUND: Tacaribe virus (TCRV) is a less biohazardous relative of the highly pathogenic clade B New World arenaviruses that cause viral hemorrhagic fever syndromes and require handling in maximum containment facilities not readily available to most researchers. AG129 type I and II interferon receptor knockout mice have been shown to be susceptible to TCRV infection, but the pathogenic mechanisms contributing to the lethal disease are unclear. METHODS: To gain insights into the pathogenesis of TCRV infection in AG129 mice, we assessed hematologic and cytokine responses during the course of infection, as well as changes in the permeability of the vascular endothelium. We also treated TCRV-challenged mice with MY-24, a compound that prevents mortality without affecting viral loads during the acute infection, and measured serum and tissue viral titers out to 40 days post-infection to determine whether the virus is ultimately cleared in recovering mice. RESULTS: We found that the development of viremia and splenomegaly precedes an elevation in white blood cells and the detection of high levels of proinflammatory mediators known to destabilize the endothelial barrier, which likely contributes to the increased vascular permeability and weight loss that was observed several days prior to when the mice generally succumb to TCRV challenge. In surviving mice treated with MY-24, viremia and liver virus titers were not cleared until 2-3 weeks post-infection, after which the mice began to recover lost weight. Remarkably, substantial viral loads were still present in the lung, spleen, brain and kidney tissues at the conclusion of the study. CONCLUSIONS: Our findings suggest that vascular leak may be a contributing factor in the demise of TCRV-infected mice, as histopathologic findings are generally mild to moderate in nature, and as evidenced with MY-24 treatment, animals can survive in the face of high viral loads.


Assuntos
Infecções por Arenaviridae/imunologia , Infecções por Arenaviridae/patologia , Arenavirus do Novo Mundo/imunologia , Arenavirus do Novo Mundo/patogenicidade , Permeabilidade Capilar , Citocinas/metabolismo , Estruturas Animais/virologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Carga Viral , Viremia/imunologia , Viremia/patologia
16.
Artigo em Russo | MEDLINE | ID: mdl-24000605

RESUMO

Analysis of data of the available literature on epidemiology of Bolivian hemorrhagic fever, manifestations of human disease, biological properties of the causative agent and development carried out abroad of means and methods of diagnostics, prophylaxis and therapy of this infection that presents a potential threat for the population and economy of the Russian Federation in case of introduction of the causative agent is presented.


Assuntos
Arenavirus do Novo Mundo/fisiologia , Febre Hemorrágica Americana/epidemiologia , Febre Hemorrágica Americana/fisiopatologia , Muridae/virologia , Vírion/fisiologia , Animais , Anticorpos Neutralizantes/sangue , Antivirais/uso terapêutico , Arenavirus do Novo Mundo/patogenicidade , Gerenciamento Clínico , Surtos de Doenças , Vetores de Doenças , Febre Hemorrágica Americana/terapia , Febre Hemorrágica Americana/virologia , Humanos , Federação Russa/epidemiologia , América do Sul/epidemiologia , Vírion/patogenicidade , Replicação Viral
17.
Mol Ecol ; 21(16): 4137-50, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22693963

RESUMO

We empirically tested the long-standing hypothesis of codivergence of New World arenaviruses (NWA) with their hosts. We constructed phylogenies for NWA and all known hosts and used them in reconciliation analyses. We also constructed a phylogenetic tree of all Sigmodontinae and Neotominae rodents and tested whether viral-host associations were phylogenetically clustered. We determined host geographical overlap to determine to what extent opportunity to switch hosts was limited by host relatedness or physical proximity. With the exception of viruses from North America, no phylogenetically codivergent pattern between NWA and their hosts was found. We found that different virus clades were clustered differently and that Clade B with members pathogenic to humans was randomly distributed across the rodent phylogeny. Furthermore, viral relatedness within Clade B was significantly explained by the geographic overlap of their hosts' ranges rather than host relatedness, indicating that they are capable of host switching opportunistically. This has important bearings on their potential to become panzootic. Together, these analyses suggest that NWA have not codiverged with their hosts and instead have evolved predominantly via host switching.


Assuntos
Arenavirus do Novo Mundo/genética , Interações Hospedeiro-Patógeno , Filogenia , Roedores/virologia , Animais , Arenavirus do Novo Mundo/patogenicidade , Análise por Conglomerados , Roedores/genética
18.
J Virol ; 84(4): 1785-91, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20007272

RESUMO

The retinoic acid-inducible gene I product (RIG-I) is a cellular sensor of RNA virus infection that regulates the cellular beta interferon (IFN-beta) response. The nucleoproteins (NP) of arenaviruses are reported to antagonize the IFN response by inhibiting interferon regulatory factor 3 (IRF-3). Here, we demonstrate that the Z proteins of four New World (NW) arenaviruses, Guanarito virus (GTOV), Junin virus (JUNV), Machupo virus (MAVC), and Sabia virus (SABV), bind to RIG-I, resulting in downregulation of the IFN-beta response. We show that expression of the four NW arenavirus Z proteins inhibits IFN-beta mRNA induction in A549 cells in response to RNA bearing 5' phosphates (5'pppRNA). NW arenavirus Z proteins interact with RIG-I in coimmunoprecipitation studies and colocalize with RIG-I. Furthermore, expression of Z proteins interferes with the interaction between RIG-I and MAVS. Z expression also impedes the nuclear factor kappa light chain enhancer of activated B cells (NF-kappaB) and IRF-3 activation. Our results indicate that NW arenavirus Z proteins, but not Z protein of the Old World (OW) arenavirus lymphocytic choriomeningitis virus (LCMV) or Lassa virus, bind to RIG-I and inhibit downstream activation of the RIG-I signaling pathway, preventing the transcriptional induction of IFN-beta.


Assuntos
Arenavirus do Novo Mundo/imunologia , Arenavirus do Novo Mundo/patogenicidade , RNA Helicases DEAD-box/metabolismo , Interferon beta/biossíntese , Proteínas Virais/metabolismo , Arenavirus do Novo Mundo/fisiologia , Sequência de Bases , Linhagem Celular , Proteína DEAD-box 58 , Humanos , Imunidade Inata , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/genética , NF-kappa B/metabolismo , Ligação Proteica , Mapeamento de Interação de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Imunológicos , Transdução de Sinais , Proteínas Virais/genética , Proteínas Virais/imunologia
19.
J Virol ; 84(2): 1176-82, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19889757

RESUMO

Clade B of the New World arenaviruses contains both pathogenic and nonpathogenic members, whose surface glycoproteins (GPs) are characterized by different abilities to use the human transferrin receptor type 1 (hTfR1) protein as a receptor. Using closely related pairs of pathogenic and nonpathogenic viruses, we investigated the determinants of the GP1 subunit that confer these different characteristics. We identified a central region (residues 85 to 221) in the Guanarito virus GP1 that was sufficient to interact with hTfR1, with residues 159 to 221 being essential. The recently solved structure of part of the Machupo virus GP1 suggests an explanation for these requirements.


Assuntos
Arenavirus do Novo Mundo/fisiologia , Arenavirus do Novo Mundo/patogenicidade , Proteínas Recombinantes de Fusão , Proteínas do Envelope Viral , Sequência de Aminoácidos , Animais , Arenavirus do Novo Mundo/classificação , Arenavirus do Novo Mundo/genética , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Receptores da Transferrina/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
20.
J Virol ; 84(19): 9947-56, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20668086

RESUMO

Arenaviruses cause severe human disease ranging from aseptic meningitis following lymphocytic choriomeningitis virus (LCMV) infection to hemorrhagic fever syndromes following infection with Guanarito virus (GTOV), Junin virus (JUNV), Lassa virus (LASV), Machupo virus (MACV), Sabia virus (SABV), or Whitewater Arroyo virus (WWAV). Cellular immunity, chiefly the CD8(+) T-cell response, plays a critical role in providing protective immunity following infection with the Old World arenaviruses LASV and LCMV. In the current study, we evaluated whether HLA class I-restricted epitopes that are cross-reactive among pathogenic arenaviruses could be identified for the purpose of developing an epitope-based vaccination approach that would cross-protect against multiple arenaviruses. We were able to identify a panel of HLA-A*0201-restricted peptides derived from the same region of the glycoprotein precursor (GPC) of LASV (GPC spanning residues 441 to 449 [GPC(441-449)]), LCMV (GPC(447-455)), JUNV (GPC(429-437)), MACV (GPC(444-452)), GTOV (GPC(427-435)), and WWAV (GPC(428-436)) that displayed high-affinity binding to HLA-A*0201 and were recognized by CD8(+) T cells in a cross-reactive manner following LCMV infection or peptide immunization of HLA-A*0201 transgenic mice. Immunization of HLA-A*0201 mice with the Old World peptide LASV GPC(441-449) or LCMV GPC(447-455) induced high-avidity CD8(+) T-cell responses that were able to kill syngeneic target cells pulsed with either LASV GPC(441-449) or LCMV GPC(447-455) in vivo and provided significant protection against viral challenge with LCMV. Through this study, we have demonstrated that HLA class I-restricted, cross-reactive epitopes exist among diverse arenaviruses and that individual epitopes can be utilized as effective vaccine determinants for multiple pathogenic arenaviruses.


Assuntos
Infecções por Arenaviridae/imunologia , Infecções por Arenaviridae/prevenção & controle , Arenavirus do Velho Mundo , Vacinas Virais/administração & dosagem , Sequência de Aminoácidos , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/virologia , Antígenos Virais/genética , Infecções por Arenaviridae/genética , Arenavirus do Novo Mundo/genética , Arenavirus do Novo Mundo/imunologia , Arenavirus do Novo Mundo/patogenicidade , Arenavirus do Velho Mundo/genética , Arenavirus do Velho Mundo/imunologia , Arenavirus do Velho Mundo/patogenicidade , Linfócitos T CD8-Positivos/imunologia , Reações Cruzadas , Citotoxicidade Imunológica , Epitopos/administração & dosagem , Epitopos/genética , Antígenos HLA-A/genética , Antígeno HLA-A2 , Humanos , Vírus Lassa/genética , Vírus Lassa/imunologia , Vírus Lassa/patogenicidade , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/patogenicidade , Camundongos , Camundongos Transgênicos , Vacinas Virais/genética , Vacinas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA