Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Med Virol ; 95(11): e29211, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37975336

RESUMEN

The emerging viruses SARS-CoV-2 and arenaviruses cause severe respiratory and hemorrhagic diseases, respectively. The production of infectious particles of both viruses and virus spread in tissues requires cleavage of surface glycoproteins (GPs) by host proprotein convertases (PCs). SARS-CoV-2 and arenaviruses rely on GP cleavage by PCs furin and subtilisin kexin isozyme-1/site-1 protease (SKI-1/S1P), respectively. We report improved luciferase-based reporter cell lines, named luminescent inducible proprotein convertase reporter cells that we employ to monitor PC activity in its authentic subcellular compartment. Using these sensor lines we screened a small compound library in high-throughput manner. We identified 23 FDA-approved small molecules, among them monensin which displayed broad activity against furin and SKI-1/S1P. Monensin inhibited arenaviruses and SARS-CoV-2 in a dose-dependent manner. We observed a strong reduction in infectious particle release upon monensin treatment with little effect on released genome copies. This was reflected by inhibition of SARS-CoV-2 spike processing suggesting the release of immature particles. In a proof of concept experiment using human precision cut lung slices, monensin potently inhibited SARS-CoV-2 infection, evidenced by reduced infectious particle release. We propose that our PC sensor pipeline is a suitable tool to identify broad-spectrum antivirals with therapeutic potential to combat current and future emerging viruses.


Asunto(s)
Arenavirus , Furina , Humanos , Furina/metabolismo , Proteínas del Envoltorio Viral/genética , Monensina/metabolismo , Monensina/farmacología , Arenavirus/genética , Arenavirus/metabolismo , Antivirales/uso terapéutico
2.
PLoS Pathog ; 19(7): e1011217, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37494374

RESUMEN

Many enveloped viruses enter host cells by fusing with acidic endosomes. The fusion activity of multiple viral envelope glycoproteins does not generally affect viral membrane permeability. However, fusion induced by the Lassa virus (LASV) glycoprotein complex (GPc) is always preceded by an increase in viral membrane permeability and the ensuing acidification of the virion interior. Here, systematic investigation of this LASV fusion phenotype using single pseudovirus tracking in live cells reveals that the change in membrane barrier function is associated with the fusogenic conformational reorganization of GPc. We show that a small-molecule fusion inhibitor or mutations that impair viral fusion by interfering with GPc refolding into the post-fusion structure prevent the increase in membrane permeability. We find that the increase in virion membrane permeability occurs early during endosomal maturation and is facilitated by virus-cell contact. This increase is observed using diverse arenavirus glycoproteins, whether presented on lentivirus-based pseudoviruses or arenavirus-like particles, and in multiple different cell types. Collectively, these results suggest that conformational changes in GPc triggered by low pH and cell factor binding are responsible for virion membrane permeabilization and acidification of the virion core prior to fusion. We propose that this viroporin-like activity may augment viral fusion and/or post-fusion steps of infection, including ribonucleoprotein release into the cytoplasm.


Asunto(s)
Arenavirus , Arenavirus/genética , Proteínas Viroporinas/metabolismo , Glicoproteínas/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Virus Lassa , Internalización del Virus
3.
PLoS One ; 16(3): e0246981, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33730017

RESUMEN

Nidoviruses and arenaviruses are the only known RNA viruses encoding a 3'-5' exonuclease domain (ExoN). The proofreading activity of the ExoN domain has played a key role in the growth of nidoviral genomes, while in arenaviruses this domain partakes in the suppression of the host innate immune signaling. Sequence and structural homology analyses suggest that these proteins have been hijacked from cellular hosts many times. Analysis of the available nidoviral ExoN sequences reveals a high conservation level comparable to that of the viral RNA-dependent RNA polymerases (RdRp), which are the most conserved viral proteins. Two highly preserved zinc fingers are present in all nidoviral exonucleases, while in the arenaviral protein only one zinc finger can be identified. This is in sharp contrast with the reported lack of zinc fingers in cellular ExoNs, and opens the possibility of therapeutic strategies in the struggle against COVID-19.


Asunto(s)
Exonucleasas/genética , Dominios Proteicos/genética , ARN Viral/genética , Proteínas Virales/genética , Secuencia de Aminoácidos , Arenavirus/genética , COVID-19/virología , Humanos , Inmunidad Innata/genética , Nidovirales/genética , Virus ARN/genética , ARN Polimerasa Dependiente del ARN/genética , SARS-CoV-2/genética , Dedos de Zinc/genética
4.
J Virol ; 93(22)2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31462569

RESUMEN

Several mammarenaviruses can cause deadly hemorrhagic fever infections in humans, with limited preventative and therapeutic measures available. Arenavirus cell entry is mediated by the viral glycoprotein (GP) complex, which consists of the stable signal peptide (SSP), the receptor-binding subunit GP1, and the transmembrane subunit GP2. The GP2 cytoplasmic tail (CT) is relatively conserved among arenaviruses and is known to interact with the SSP to regulate GP processing and membrane fusion, but its biological role in the context of an infectious virus has not been fully characterized. Using a Pichinde virus (PICV) GP expression vector and a PICV reverse genetics system, we systematically characterized the functional roles of 12 conserved residues within the GP2 CT in GP processing, trafficking, assembly, and fusion, as well as in viral replication. Except for P478A and K505A R508A, alanine substitutions at conserved residues abolished GP processing and membrane fusion in plasmid-transfected cells. Six invariant H and C residues and W503 are essential for viral replication, as evidenced by the fact that their mutant viruses could not be rescued. Both P480A and R482A mutant viruses were rescued, grew similarly to wild-type (WT) virus, and produced evidently processed GP1 and GP2 subunits in virus-infected cells, despite the fact that the same mutations abolished GP processing and membrane fusion in a plasmid-based protein expression system, illustrating the importance of using an infectious-virus system for analyzing viral glycoprotein function. In summary, our results demonstrate an essential biological role of the GP2 CT in arenavirus replication and suggest it as a potential novel target for developing antivirals and/or attenuated viral vaccine candidates.IMPORTANCE Several arenaviruses, such as Lassa virus (LASV), can cause severe and lethal hemorrhagic fever diseases with high mortality and morbidity, for which no FDA-approved vaccines or therapeutics are available. Viral entry is mediated by the arenavirus GP complex, which consists of the stable signal peptide (SSP), the receptor-binding subunit GP1, and the transmembrane subunit GP2. The cytoplasmic tail (CT) of GP2 is highly conserved among arenaviruses, but its functional role in viral replication is not completely understood. Using a reverse genetics system of a prototypic arenavirus, Pichinde virus (PICV), we show that the GP2 CT contains certain conserved residues that are essential for virus replication, implicating it as a potentially good target for developing antivirals and live-attenuated viral vaccines against deadly arenavirus pathogens.


Asunto(s)
Glicoproteínas/metabolismo , Virus Pichinde/genética , Proteínas del Envoltorio Viral/genética , Células A549 , Sustitución de Aminoácidos/genética , Animales , Arenaviridae , Infecciones por Arenaviridae/genética , Infecciones por Arenaviridae/metabolismo , Arenavirus/genética , Arenavirus/metabolismo , Línea Celular , Chlorocebus aethiops , Glicoproteínas/genética , Células HEK293 , Humanos , Fusión de Membrana/genética , Mutación/genética , Virus Pichinde/metabolismo , Señales de Clasificación de Proteína/genética , Células Vero , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Replicación Viral
5.
Methods Mol Biol ; 1604: 169-178, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28986832

RESUMEN

Arenaviruses, such as Lassa virus (LASV) and Pichindé virus (PICV), are enveloped viruses with a bi-segmented ambisense RNA genome. The large (L) genomic segment encodes the Z matrix protein and the L RNA-dependent RNA polymerase, whereas the small (S) genomic segment encodes the nucleoprotein (NP) and the glycoprotein precursor complex (GPC). GPC is processed by signal peptidase in the endoplasmic reticulum into the stable signal peptide (SSP) and GP1/GP2, which is further cleaved by the Golgi-resident subtilisin kexin isozyme-1 (SKI-1)/site-1 protease (S1P) into the cellular receptor-recognition subunit GP1 and the transmembrane subunit GP2, which helps promote the membrane fusion reaction to allow virus entry into the cell. This article describes assays to assess PICV GPC expression, proteolytic processing, fusion function, and GPC-mediated virus-like particle (VLP) entry into cells under tissue-culture conditions.


Asunto(s)
Arenavirus/metabolismo , Bioensayo/métodos , Glicoproteínas/metabolismo , Animales , Arenavirus/genética , Glicoproteínas/genética , Humanos , Virus Lassa/genética , Virus Lassa/metabolismo , Virus Pichinde/genética , Virus Pichinde/metabolismo , Proproteína Convertasas/genética , Proproteína Convertasas/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo
6.
J Virol ; 90(22): 10390-10397, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27630230

RESUMEN

Arenaviruses can cause lethal hemorrhagic fevers in humans with few preventative and therapeutic measures. The arenaviral glycoprotein stable signal peptide (SSP) is unique among signal peptides in that it is an integral component of the mature glycoprotein complex (GPC) and plays important roles not only in GPC expression and processing but also in the membrane fusion process during viral entry. Using the Pichinde virus (PICV) reverse genetics system, we analyzed the effects of alanine substitutions at many conserved residues within the SSP on viral replication in cell culture and in a guinea pig infection model. Our data showed that the K33A, F49A, and C57A mutations abolished GPC-mediated cell entry and therefore could not allow for the generation of viable recombinant viruses, demonstrating that these residues are essential for the PICV life cycle. The G2A mutation caused a marked reduction of cell entry at the membrane fusion step, and while this mutant virus was viable, it was significantly attenuated in vitro and in vivo The N20A mutation also reduced membrane fusion activity and viral virulence in guinea pigs, but it did not significantly affect cell entry or viral growth in cell culture. Two other mutations (N37A and R55A) did not affect membrane fusion or viral growth in vitro but significantly reduced viral virulence in vivo Taken together, our data suggest that the GPC SSP plays an essential role in mediating viral entry and also contributes to viral virulence in vivo IMPORTANCE: Several arenaviruses, such as Lassa fever virus, can cause severe and lethal hemorrhagic fever diseases with high mortality and morbidity, and no FDA-approved vaccines or therapies are currently available. Viral entry into cells is mediated by arenavirus GPC that consists of an SSP, the receptor-binding GP1, and transmembrane GP2 protein subunits. Using a reverse genetics system of a prototypic arenavirus, Pichinde virus (PICV), we have shown for the first time in the context of virus infections of cell culture and of guinea pigs that the SSP plays an essential role in mediating the membrane fusion step as well as in other yet-to-be-determined processes during viral infection. Our study provides important insights into the biological roles of GPC SSP and implicates it as a good target for the development of antivirals against deadly human arenavirus pathogens.


Asunto(s)
Glicoproteínas/genética , Virus Pichinde/genética , Señales de Clasificación de Proteína/genética , Virulencia/genética , Células A549 , Animales , Infecciones por Arenaviridae/virología , Arenavirus/genética , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Replicación del ADN/genética , Cobayas , Células HEK293 , Humanos , Fusión de Membrana/genética , Mutación/genética , Subunidades de Proteína/genética , Células Vero , Proteínas del Envoltorio Viral/genética , Internalización del Virus , Replicación Viral/genética
7.
Curr Top Microbiol Immunol ; 392: 231-76, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26472215

RESUMEN

The family Arenaviridae currently comprises over 20 viral species, each of them associated with a main rodent species as the natural reservoir and in one case possibly phyllostomid bats. Moreover, recent findings have documented a divergent group of arenaviruses in captive alethinophidian snakes. Human infections occur through mucosal exposure to aerosols or by direct contact of abraded skin with infectious materials. Arenaviruses merit interest both as highly tractable experimental model systems to study acute and persistent infections and as clinically important human pathogens including Lassa (LASV) and Junin (JUNV) viruses, the causative agents of Lassa and Argentine hemorrhagic fevers (AHFs), respectively, for which there are no FDA-licensed vaccines, and current therapy is limited to an off-label use of ribavirin (Rib) that has significant limitations. Arenaviruses are enveloped viruses with a bi-segmented negative strand (NS) RNA genome. Each genome segment, L (ca 7.3 kb) and S (ca 3.5 kb), uses an ambisense coding strategy to direct the synthesis of two polypeptides in opposite orientation, separated by a noncoding intergenic region (IGR). The S genomic RNA encodes the virus nucleoprotein (NP) and the precursor (GPC) of the virus surface glycoprotein that mediates virus receptor recognition and cell entry via endocytosis. The L genome RNA encodes the viral RNA-dependent RNA polymerase (RdRp, or L polymerase) and the small (ca 11 kDa) RING finger protein Z that has functions of a bona fide matrix protein including directing virus budding. Arenaviruses were thought to be relatively stable genetically with intra- and interspecies amino acid sequence identities of 90-95 % and 44-63 %, respectively. However, recent evidence has documented extensive arenavirus genetic variability in the field. Moreover, dramatic phenotypic differences have been documented among closely related LCMV isolates. These data provide strong evidence of viral quasispecies involvement in arenavirus adaptability and pathogenesis. Here, we will review several aspects of the molecular biology of arenaviruses, phylogeny and evolution, and quasispecies dynamics of arenavirus populations for a better understanding of arenavirus pathogenesis, as well as for the development of novel antiviral strategies to combat arenavirus infections.


Asunto(s)
Infecciones por Arenaviridae/virología , Arenavirus/genética , Evolución Molecular , Animales , Antivirales/farmacología , Infecciones por Arenaviridae/tratamiento farmacológico , Arenavirus/clasificación , Arenavirus/efectos de los fármacos , Arenavirus/fisiología , Variación Genética , Genoma Viral , Humanos , Filogenia , Replicación Viral
8.
J Vet Diagn Invest ; 27(2): 245-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25776548

RESUMEN

A captive bred red tail boa (Boa constrictor constrictor) was presented with a large intraoral mass originating from the buccal gingiva, attached to the right dentary teeth row. Based on the clinical features and histological examination, the diagnosis of a peripheral odontogenic fibromyxoma was made. Sections of liver biopsies and circulating lymphocytes contained relatively few eosinophilic intracytoplasmic inclusion bodies, indistinguishable from those observed in inclusion body disease-affected snakes. Inclusion bodies were not observed in cells comprising the neoplastic mass. Using reverse transcription polymerase chain reaction (RT-PCR), arenavirus was detected in the neoplastic tissue. Two years after surgical removal of the mass, recurrence of the neoplastic lesion was observed. Numerous large inclusion body disease inclusions were abundantly present in the neoplastic cells of the recurrent fibromyxoma. Sections of liver biopsies and circulating lymphocytes contained relatively few intracytoplasmic inclusions. The RT-PCR revealed the presence of arenavirus in blood, a liver biopsy, and neoplastic tissue. The present case describes the co-occurrence of an arenavirus infection and an odontogenic fibromyxoma in a red tail boa.


Asunto(s)
Infecciones por Arenaviridae/veterinaria , Arenavirus/aislamiento & purificación , Boidae , Fibromatosis Gingival/veterinaria , Cuerpos de Inclusión Viral/patología , Animales , Infecciones por Arenaviridae/diagnóstico , Infecciones por Arenaviridae/virología , Arenavirus/genética , Biopsia , ADN Viral/análisis , Diagnóstico Diferencial , Fibromatosis Gingival/diagnóstico , Fibromatosis Gingival/virología , Masculino , Reacción en Cadena de la Polimerasa/veterinaria
9.
Arch Virol ; 158(9): 1895-905, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23553456

RESUMEN

The Lassa virus nucleoprotein (NP) is a multifunctional protein that plays an essential role in many aspects of the viral life cycle, including RNA encapsidation, viral transcription and replication, recruitment of ribonucleoprotein complexes to viral budding sites, and inhibition of the host cell interferon response. While it is known that NP is capable of forming oligomers, both the oligomeric state of NP in mammalian cells and the significance of NP oligomerization for its various functions remain unclear. Here, we demonstrate that Lassa virus NP solely forms trimers upon expression in mammalian cells. Using a minigenome assay we show that mutants that are not able to form stable trimers are no longer functional during transcription and/or replication of the minigenome, indicating that NP trimerization is essential for transcription and/or replication of the viral genome. However, mutations leading to destabilization of the NP trimer did not impact the incorporation of NP into virus-like particles or its ability to suppress interferon-induced gene expression, two important functions of arenavirus NP.


Asunto(s)
Arenavirus/metabolismo , Nucleoproteínas/metabolismo , Secuencia de Aminoácidos , Arenavirus/genética , Línea Celular Tumoral , Células HEK293 , Humanos , Virus Lassa/genética , Virus Lassa/metabolismo , Datos de Secuencia Molecular , Nucleoproteínas/química , Nucleoproteínas/genética , Multimerización de Proteína , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral
10.
Methods Mol Biol ; 939: 253-61, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23192551

RESUMEN

One of the challenges of genetic data analysis is to combine information from sources that are distributed around the world and accessible through a wide array of different methods and interfaces. The HIV database and its footsteps, the hepatitis C virus (HCV) and hemorrhagic fever virus (HFV) databases, have made it their mission to make different data types easily available to their users. This involves a large amount of behind-the-scenes processing, including quality control and analysis of the sequences and their annotation. Gene and protein sequences are distilled from the sequences that are stored in GenBank; to this end, both submitter annotation and script-generated sequences are used. Alignments of both nucleotide and amino acid sequences are generated, manually curated, distilled into an alignment model, and regenerated in an iterative cycle that results in ever better new alignments. Annotation of epidemiological and clinical information is parsed, checked, and added to the database. User interfaces are updated, and new interfaces are added based upon user requests. Vital for its success, the database staff are heavy users of the system, which enables them to fix bugs and find opportunities for improvement. In this chapter we describe some of the infrastructure that keeps these heavily used analysis platforms alive and vital after nearly 25 years of use. The database/analysis platforms described in this chapter can be accessed at http://hiv.lanl.gov http://hcv.lanl.gov http://hfv.lanl.gov.


Asunto(s)
Biología Computacional/métodos , Genoma Viral , Genómica/métodos , Gestión del Conocimiento , Arenavirus/clasificación , Arenavirus/genética , Bases de Datos Genéticas , VIH/clasificación , VIH/genética , Hepacivirus/clasificación , Hepacivirus/genética , Alineación de Secuencia , Análisis de Secuencia de ARN , Programas Informáticos
11.
J Virol ; 86(11): 6138-45, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22438561

RESUMEN

The arenavirus envelope glycoprotein (GPC) retains a stable signal peptide (SSP) as an essential subunit in the mature complex. The 58-amino-acid residue SSP comprises two membrane-spanning hydrophobic regions separated by a short ectodomain loop that interacts with the G2 fusion subunit to promote pH-dependent membrane fusion. Small-molecule compounds that target this unique SSP-G2 interaction prevent arenavirus entry and infection. The interaction between SSP and G2 is sensitive to the phylogenetic distance between New World (Junín) and Old World (Lassa) arenaviruses. For example, heterotypic GPC complexes are unable to support virion entry. In this report, we demonstrate that the hybrid GPC complexes are properly assembled, proteolytically cleaved, and transported to the cell surface but are specifically defective in their membrane fusion activity. Chimeric SSP constructs reveal that this incompatibility is localized to the first transmembrane segment of SSP (TM1). Genetic changes in TM1 also affect sensitivity to small-molecule fusion inhibitors, generating resistance in some cases and inhibitor dependence in others. Our studies suggest that interactions of SSP TM1 with the transmembrane domain of G2 may be important for GPC-mediated membrane fusion and its inhibition.


Asunto(s)
Arenavirus/fisiología , Glicoproteínas/metabolismo , Señales de Clasificación de Proteína , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Secuencia de Aminoácidos , Animales , Arenavirus/genética , Chlorocebus aethiops , Glicoproteínas/genética , Modelos Moleculares , Datos de Secuencia Molecular , Células Vero , Proteínas del Envoltorio Viral/genética
12.
J Virol ; 86(9): 4935-46, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22357276

RESUMEN

A crucial step in the life cycle of arenaviruses is the biosynthesis of the mature fusion-active viral envelope glycoprotein (GP) that is essential for virus-host cell attachment and entry. The maturation of the arenavirus GP precursor (GPC) critically depends on proteolytic processing by the cellular proprotein convertase (PC) subtilisin kexin isozyme-1 (SKI-1)/site-1 protease (S1P). Here we undertook a molecular characterization of the SKI-1/S1P processing of the GPCs of the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) and the pathogenic Lassa virus (LASV). Previous studies showed that the GPC of LASV undergoes processing in the endoplasmic reticulum (ER)/cis-Golgi compartment, whereas the LCMV GPC is cleaved in a late Golgi compartment. Herein we confirm these findings and provide evidence that the SKI-1/S1P recognition site RRLL, present in the SKI-1/S1P prodomain and LASV GPC, but not in the LCMV GPC, is crucial for the processing of the LASV GPC in the ER/cis-Golgi compartment. Our structure-function analysis revealed that the cleavage of arenavirus GPCs, but not cellular substrates, critically depends on the autoprocessing of SKI-1/S1P, suggesting differences in the processing of cellular and viral substrates. Deletion mutagenesis showed that the transmembrane and intracellular domains of SKI-1/S1P are dispensable for arenavirus GPC processing. The expression of a soluble form of the protease in SKI-I/S1P-deficient cells resulted in the efficient processing of arenavirus GPCs and rescued productive virus infection. However, exogenous soluble SKI-1/S1P was unable to process LCMV and LASV GPCs displayed at the surface of SKI-I/S1P-deficient cells, indicating that GPC processing occurs in an intracellular compartment. In sum, our study reveals important differences in the SKI-1/S1P processing of viral and cellular substrates.


Asunto(s)
Arenavirus/metabolismo , Proproteína Convertasas/metabolismo , Serina Endopeptidasas/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Secuencias de Aminoácidos , Animales , Arenavirus/genética , Línea Celular , Membrana Celular/metabolismo , Cricetinae , Retículo Endoplásmico/metabolismo , Orden Génico , Aparato de Golgi/metabolismo , Humanos , Proproteína Convertasas/química , Estructura Terciaria de Proteína , Serina Endopeptidasas/química , Solubilidad , Especificidad por Sustrato , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética
13.
J Virol ; 86(8): 4578-85, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22345463

RESUMEN

Several arenaviruses, chiefly Lassa virus (LASV), cause hemorrhagic fever (HF) disease in humans and pose a significant public health concern in regions where they are endemic. On the other hand, evidence indicates that the globally distributed prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) is a neglected human pathogen. The phosphatidylinositol 3-kinase (PI3K)/Akt pathway participates in many cellular processes, including cell survival and differentiation, and also has been shown to play important roles in different steps of the life cycles of a variety of viruses. Here we report that the inhibition of the PI3K/Akt pathway inhibited budding and to a lesser extent RNA synthesis, but not cell entry, of LCMV. Accordingly, BEZ-235, a PI3K inhibitor currently in cancer clinical trials, inhibited LCMV multiplication in cultured cells. These findings, together with those previously reported for Junin virus (JUNV), indicate that targeting the PI3K/Akt pathway could represent a novel antiviral strategy to combat human-pathogenic arenaviruses.


Asunto(s)
Arenavirus/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Liberación del Virus , Animales , Arenavirus/efectos de los fármacos , Arenavirus/genética , Línea Celular , Chlorocebus aethiops , Cromonas/farmacología , Regulación Viral de la Expresión Génica/efectos de los fármacos , Humanos , Imidazoles/farmacología , Virus Lassa/efectos de los fármacos , Virus Lassa/genética , Virus Lassa/metabolismo , Virus de la Coriomeningitis Linfocítica/efectos de los fármacos , Virus de la Coriomeningitis Linfocítica/genética , Virus de la Coriomeningitis Linfocítica/metabolismo , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Quinolinas/farmacología , ARN Viral/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Internalización del Virus/efectos de los fármacos , Liberación del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
14.
Antimicrob Agents Chemother ; 55(10): 4631-8, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21825302

RESUMEN

Members of the Arenaviridae family are a threat to public health and can cause meningitis and hemorrhagic fever, and yet treatment options remain limited by a lack of effective antivirals. In this study, we found that peptide-conjugated phosphorodiamidate morpholino oligomers (PPMO) complementary to viral genomic RNA were effective in reducing arenavirus replication in cell cultures and in vivo. PPMO complementary to the Junín virus genome were designed to interfere with viral RNA synthesis or translation or both. However, only PPMO designed to potentially interfere with translation were effective in reducing virus replication. PPMO complementary to sequences that are highly conserved across the arenaviruses and located at the 5' termini of both genomic segments were effective against Junín virus, Tacaribe virus, Pichinde virus, and lymphocytic choriomeningitis virus (LCMV)-infected cell cultures and suppressed viral titers in the livers of LCMV-infected mice. These results suggest that arenavirus 5' genomic termini represent promising targets for pan-arenavirus antiviral therapeutic development.


Asunto(s)
Antivirales/farmacología , Arenavirus/efectos de los fármacos , Morfolinos/farmacología , Péptidos/farmacología , Animales , Infecciones por Arenaviridae/tratamiento farmacológico , Infecciones por Arenaviridae/virología , Arenavirus/genética , Arenavirus/crecimiento & desarrollo , Arenavirus del Nuevo Mundo/efectos de los fármacos , Línea Celular , Chlorocebus aethiops , Genoma Viral , Virus Junin/efectos de los fármacos , Virus de la Coriomeningitis Linfocítica/efectos de los fármacos , Ratones , Pruebas de Sensibilidad Microbiana , Virus Pichinde/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , ARN Viral/genética , Células Vero , Replicación Viral/efectos de los fármacos
15.
Virology ; 417(1): 18-26, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21612810

RESUMEN

A crucial step in the arenavirus life cycle is the proteolytic processing of the viral envelope glycoprotein precursor (GPC) by the cellular proprotein convertase (PC) subtilisin kexin isozyme-1 (SKI-1)/site-1 protease (S1P). Here we conducted a systematic and quantitative analysis of SKI-1/S1P processing of peptides derived from the recognition sites of GPCs of different Old World and New World arenaviruses. We found that SKI-1/S1P showed a strong preference for arenaviral sequences resembling its autoprocessing sites, which are recurrent motifs in arenaviral GPCs. The African arenaviruses Lassa, Mobala, and Mopeia resemble the SKI-1/S1P autoprocessing C-site, whereas sequences derived from Clade B New World viruses Junin and Tacaribe have similarities to the autoprocessing B-site. In contrast, analogous peptides derived from cellular SKI-1/S1P substrates were remarkably poor substrates. The data suggest that arenavirus GPCs evolved to mimic SKI-1/S1P autoprocessing sites, likely ensuring efficient cleavage and perhaps avoiding competition with SKI-1/S1P's cellular substrates.


Asunto(s)
Arenavirus/metabolismo , Proproteína Convertasas/metabolismo , Serina Endopeptidasas/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Arenavirus/genética , Células HEK293 , Humanos , Proproteína Convertasas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina Endopeptidasas/genética , Proteínas del Envoltorio Viral/genética
16.
J Biol Chem ; 286(8): 6108-16, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21159780

RESUMEN

Arenavirus RNA genomes are initiated by a "prime and realign" mechanism, such that the initiating GTP is found as a single unpaired (overhanging) nucleotide when the complementary genome ends anneal to form double-stranded (ds) RNA panhandle structures. dsRNAs modeled on these structures do not induce interferon (IFN), as opposed to blunt-ended (5' ppp)dsRNA. This study examines whether these viral structures can also act as decoys, by trapping RIG-I in inactive dsRNA complexes. We examined the ability of various dsRNAs to activate the RIG-I ATPase (presumably a measure of helicase translocation on dsRNA) relative to their ability to induce IFN. We found that there is no simple relationship between these two properties, as if RIG-I can translocate on short dsRNAs without inducing IFN. Moreover, we found that (5' ppp)dsRNAs with a single unpaired 5' ppp-nucleotide can in fact competitively inhibit the ability of blunt-ended (5' ppp)dsRNAs to induce IFN when co-transfected into cells and that this inhibition is strongly dependent on the presence of the 5' ppp. In contrast, (5' ppp)dsRNAs with a single unpaired 5' ppp-nucleotide does not inhibit poly(I-C)-induced IFN activation, which is independent of the presence of a 5' ppp group.


Asunto(s)
Arenavirus/metabolismo , ARN Helicasas DEAD-box/metabolismo , Genoma Viral/fisiología , ARN Bicatenario/metabolismo , ARN Viral/metabolismo , Arenavirus/genética , Línea Celular , Proteína 58 DEAD Box , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Humanos , Inductores de Interferón/farmacología , Interferones/biosíntesis , Interferones/genética , Poli I-C/farmacología , ARN Bicatenario/genética , ARN Viral/genética , Receptores Inmunológicos
17.
Rev. Soc. Bras. Med. Trop ; 43(3): 229-233, May-June 2010. ilus, mapas, tab
Artículo en Inglés | LILACS | ID: lil-548514

RESUMEN

INTRODUCTION: Arenavirus hemorrhagic fever is a severe emerging disease. METHODS: Considering that the levels of antibodies against arenavirus in the Brazilian population are completely unknown, we have standardized an ELISA test for detecting IgG antibodies using a recombinant nucleoprotein from the Junin virus as the antigen. This protein was obtained by inserting the gene of the Junin virus nucleoprotein into the genome of Autographa californica nucleopolyhedrovirus, using the Bac-to-Bac baculovirus expression system. This recombinant baculovirus was used to infect S. frugiperda cells (SF9). RESULTS: The infection resulted in synthesis of high concentrations of recombinant protein. This protein was detected on 12.5 percent polyacrylamide gel and by means of Western blot. Using the standardized ELISA test, 343 samples from the population of Nova Xavantina were analyzed. We observed that 1.4 percent of the serum samples (five samples) presented antibody titers against arenavirus. CONCLUSIONS: These results show the population studied may present exposure to arenavirus infection.


INTRODUÇÃO: A febre hemorrágica por Arenavirus é uma severa doença emergente. MÉTODOS: Considerando que os níveis de anticorpos contra Arenavirus na população brasileira é totalmente desconhecido, nos padronizamos um teste de ELISA para detecção de anticorpos IgG usando uma nucleoproteína recombinante do vírus Junin como antígeno. Esta proteína foi obtida pela inserção do gene da nucleoproteína do vírus Junin no genoma do vírus Autographa californica nucleopolyhedrovirus, utilizando o sistema de expressão em Baculovírus, Bac-To-Bac. Este baculovirus recombinante foi utilizado para infecção de células de S. frugiperda (Sf9). RESULTADOS: A infecção resultou na produção de altas concentrações de proteína recombinante. Esta proteína foi detectada em gel de poliacrilamida 12,5 por cento, e em Western blot. Utilizando o teste de ELISA padronizado, foram analizadas 343 amostras provenientes da população de Nova Xavantina. Observamos que 1,4 por cento dos soros (5 amostras) apresentavam títulos de anticorpos contra arenavírus. CONCLUSÕES: Estes resultados sugerem que a população estudada pode estar sendo exposta a infecções por arenavírus.


Asunto(s)
Adulto , Femenino , Humanos , Masculino , Anticuerpos Antivirales/sangre , Antígenos Virales/inmunología , Infecciones por Arenaviridae/diagnóstico , Arenavirus/inmunología , Ensayo de Inmunoadsorción Enzimática/normas , Virus Junin/inmunología , Arenavirus/genética , Brasil , Ensayo de Inmunoadsorción Enzimática/métodos , Inmunoglobulina G/sangre , Virus Junin/genética , Nucleoproteínas/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología
19.
Virology ; 321(1): 134-43, 2004 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-15033572

RESUMEN

Data from human studies and animal experiments indicate a dominant role of T-cells over antibodies in controlling acute Lassa virus infection and providing immunity to reinfection. Knowledge of the epitopes recognized by T-cells may therefore be crucial to the development of a recombinant Lassa virus vaccine. In order to study human T-cell reactivity to the most conserved structural protein of Lassa virus, the glycoprotein 2 (GP2), seven GP2-specific CD4+ T-cell clones (TCCs) were generated from the lymphocytes of a Lassa antibody positive individual. All TCC displayed high specific proliferation, showed DR-restriction, and produced IFN-gamma upon stimulation with recombinant GP2. The epitope of four of the clones was localized to a short stretch of 13 amino acids located in the N-terminal part of GP2 (aa 289-301, numbering according to sequence of GPC). This epitope is conserved in all strains of Lassa virus and lymphocytic choriomeningitis virus (LCMV), shows >90% similarity in all New World arenaviruses of clade B, and overlaps with the proposed fusion domain of GP2. Peptides with conservative aa exchanges, as they naturally occur in the epitope 289-301 of the Old World arenavirus Mopeia and some New World arenaviruses, continued to effectively stimulate the Lassa-GP2-specific T-cell clones tested. The finding of a human T-helper cell epitope, which is highly conserved between Old and New World arenaviruses, is of importance for the design of arenavirus vaccines.


Asunto(s)
Arenavirus/inmunología , Linfocitos T CD4-Positivos/inmunología , Epítopos de Linfocito T/inmunología , Glicoproteínas/inmunología , Proteínas del Envoltorio Viral/inmunología , Secuencia de Aminoácidos , Arenavirus/genética , Donantes de Sangre , División Celular , Células Clonales , Mapeo Epitopo , Epítopos de Linfocito T/genética , Femenino , Glicoproteínas/genética , Humanos , Virus Lassa/genética , Virus Lassa/inmunología , Masculino , Datos de Secuencia Molecular , Péptidos/genética , Péptidos/farmacología , Alineación de Secuencia , Especificidad de la Especie , Proteínas del Envoltorio Viral/genética
20.
Proc Natl Acad Sci U S A ; 100(22): 12978-83, 2003 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-14563923

RESUMEN

By using a reverse genetics system that is based on the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV), we have identified the arenavirus small RING finger Z protein as the main driving force of virus budding. Both LCMV and Lassa fever virus (LFV) Z proteins exhibited self-budding activity, and both substituted efficiently for the late domain that is present in the Gag protein of Rous sarcoma virus. LCMV and LFV Z proteins contain proline-rich motifs that are characteristic of late domains. Mutations in the PPPY motif of LCMV Z severely impaired the formation of virus-like particles. LFV Z contains two different proline-rich motifs, PPPY and PTAP, which are separated by eight amino acids. Mutational analysis revealed that both motifs are required for efficient LFV Z-mediated budding. Both LCMV and LFV Z proteins recruited to the plasma membrane Tsg101, which is a component of the class E vacuolar protein sorting machinery that has been implicated in budding of HIV and Ebola virus. Targeting of Tsg101 by RNA interference caused a strong reduction in Z-mediated budding. These results indicate that Z is the arenavirus functional counterpart of the matrix proteins found in other negative strand enveloped RNA viruses. Moreover, members of the vacuolar protein sorting pathway appear to play an important role in arena-virus budding. These findings open possibilities for antiviral strategies to combat LFV and other hemorrhagic fever arenaviruses.


Asunto(s)
Arenavirus/crecimiento & desarrollo , Proteínas Sanguíneas/fisiología , Proteínas Portadoras/fisiología , Virus de la Coriomeningitis Linfocítica/crecimiento & desarrollo , Proteínas Virales/fisiología , Secuencia de Aminoácidos , Animales , Arenavirus/genética , Proteínas Sanguíneas/genética , Células COS , Proteínas Portadoras/genética , Línea Celular , Chlorocebus aethiops , Secuencia Conservada , Cricetinae , Riñón , Virus de la Coriomeningitis Linfocítica/genética , Datos de Secuencia Molecular , Plásmidos , ARN Viral/genética , ARN Viral/aislamiento & purificación , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transfección , Proteínas Virales/genética , Dedos de Zinc
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA