Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Struct Funct ; 45(2): 155-163, 2020 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-33191384

RESUMO

The smallest arenavirus gene product, Z protein, plays critical roles in the virus life cycle. Z is the major driving force of budding and particle production because of a unique property that defines self-assembly. In addition to the roles in budding, Z also participates in the suppression of type I interferon production to evade host antiviral immunity. Therefore, Z and its assembled form are an attractive drug target for arenaviral hemorrhagic fever, such as Lassa fever. Here, we developed a biosensor that enabled the evaluation of the prototype arenavirus, lymphocytic choriomeningitis virus (LCMV), Z assembly using the principle of Förster resonance energy transfer (FRET). This FRET biosensor consisted of three tandem Z that were sandwiched between super-enhanced cyan-emitting fluorescent protein and variant of a yellow-emitting mutant of green fluorescent protein so that Z-Z intermolecular binding via the really interesting new gene finger domain increased the emission ratio. To identify novel anti-arenavirus compounds, the FRET biosensor was employed to screen the PathogenBox400 for inhibitors of Z assembly in a 96-well plate format. The assay performed well, with a Z'-factor of 0.89, and identified two compounds that decreased the emission ratio of the FRET biosensor in a dose-dependent manner. Of them, the compound, 5,6,7,8-tetrahydro-7-(benzyl)-pyrido[4',3':4,5]thieno[2,3-d]pyrimidin-2,4-diamine, was found to significantly inhibit LCMV propagation in infected cells. Thereby, the present study demonstrated that a novel FRET biosensor incorporating Z assembly built on FRET and named Zabton, was a valuable screening tool to identify anti-arenavirus compounds in the context of inhibition of Z assembly.Key words: Arenavirus, Förster resonance energy transfer, anti-viral drugs, Z protein.


Assuntos
Antivirais , Arenavirus/fisiologia , Técnicas Biossensoriais , Transferência Ressonante de Energia de Fluorescência , Proteínas Virais/metabolismo , Montagem de Vírus/efeitos dos fármacos , Antivirais/química , Antivirais/farmacologia , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Células HeLa , Humanos
2.
PLoS Pathog ; 13(1): e1006073, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28076397

RESUMO

Many emerging infections are RNA virus spillovers from animal reservoirs. Reservoir identification is necessary for predicting the geographic extent of infection risk, but rarely are taxonomic levels below the animal species considered as reservoir, and only key circumstances in nature and methodology allow intrinsic virus-host associations to be distinguished from simple geographic (co-)isolation. We sampled and genetically characterized in detail a contact zone of two subtaxa of the rodent Mastomys natalensis in Tanzania. We find two distinct arenaviruses, Gairo and Morogoro virus, each spatially confined to a single M. natalensis subtaxon, only co-occurring at the contact zone's centre. Inter-subtaxon hybridization at this centre and a continuum of quality habitat for M. natalensis show that both viruses have the ecological opportunity to spread into the other substaxon's range, but do not, strongly suggesting host-intrinsic barriers. Such barriers could explain why human cases of another M. natalensis-borne arenavirus, Lassa virus, are limited to West Africa.


Assuntos
Arenavirus/classificação , Arenavirus/metabolismo , Reservatórios de Doenças/virologia , Murinae/virologia , Doenças dos Roedores/virologia , Animais , Arenavirus/fisiologia , Humanos , Febre Lassa/virologia , Vírus Lassa/fisiologia , Filogeografia , Especificidade da Espécie , Tanzânia
3.
J Gen Virol ; 99(2): 187-193, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29393022

RESUMO

We report the development of recombinant New World (Junín; JUNV) and Old World (lymphocytic choriomeningitis virus; LCMV) mammarenaviruses that encode an HA-tagged matrix protein (Z). These viruses permit the robust affinity purification of Z from infected cells or virions, as well as the detection of Z by immunofluorescent microscopy. Importantly, the HA-tagged viruses grow with wild-type kinetics in a multi-cycle growth assay. Using these viruses, we report a novel description of JUNV Z localization in infected cells, as well as the first description of colocalization between LCMV Z and the GTPase Rab5c. This latter result, when combined with our previous findings that LCMV genome and glycoprotein also colocalize with Rab5c, suggest that LCMV may target Rab5c-positive membranes for preassembly of virus particles prior to budding. The recombinant viruses reported here will provide the field with new tools to better study Z protein functionality and identify key Z protein interactions with host machinery.


Assuntos
Arenavirus/fisiologia , Proteínas de Transporte/metabolismo , Epitopos/imunologia , GTP Fosfo-Hidrolases/metabolismo , Interações Hospedeiro-Patógeno , Vírus da Coriomeningite Linfocítica/fisiologia , Células A549 , Arenavirus/imunologia , Proteínas de Transporte/genética , Endossomos/metabolismo , Endossomos/virologia , GTP Fosfo-Hidrolases/genética , Genes Reporter , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/virologia , Peptídeos e Proteínas de Sinalização Intracelular , Vírus da Coriomeningite Linfocítica/imunologia , Microscopia de Fluorescência , Montagem de Vírus
4.
Uirusu ; 68(1): 51-62, 2018.
Artigo em Japonês | MEDLINE | ID: mdl-31105135

RESUMO

Arenavirus is a genetic term for viruses belonging to the family Arenaviridae and is presented from lymphocytic choriomeningitis virus (LCMV), which shows almost no pathogenicity to humans, to Lassa virus, Junin virus, Machupo virus, Chapare virus, Lujo virus, Sabia virus, and Guanarito virus, which shows high pathogenicity to humans. These viruses except for LCMV are risk group 4 pathogens specified by World Health Organization. Based on this designation, it is designated as Class I pathogens in Japan. Although there have been no reports excluding one imported case of the Lassa fever patient, it is not surprising whenever imported cases occur in our country. Considering the disease severity and mortality rate, it is an urgent matter to develop vaccines and therapeutic drugs in endemic areas, and maintenances of these are also important in countries other than endemic areas. However, basic research on highly pathogenic arenavirus infections and development of therapeutic drugs are not easily progressed, because handling in highly safe research facilities is indispensable. In this article, we will outline the current knowledge from the recent basic research on arenavirus to the development situation of antivirals against arenaviruses.


Assuntos
Antivirais , Infecções por Arenaviridae/tratamento farmacológico , Infecções por Arenaviridae/virologia , Arenavirus/classificação , Arenavirus/patogenicidade , Descoberta de Drogas , África Ocidental/epidemiologia , Infecções por Arenaviridae/epidemiologia , Infecções por Arenaviridae/prevenção & controle , Arenavirus/genética , Arenavirus/fisiologia , Surtos de Doenças , Descoberta de Drogas/tendências , Genoma Viral/genética , Humanos , Pesquisa/tendências , Transcrição Gênica , Vacinas Virais , Vírion
5.
PLoS Pathog ; 11(10): e1005220, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26513362

RESUMO

Hemorrhagic fever viruses, including the filoviruses (Ebola and Marburg) and arenaviruses (Lassa and Junín viruses), are serious human pathogens for which there are currently no FDA approved therapeutics or vaccines. Importantly, transmission of these viruses, and specifically late steps of budding, critically depend upon host cell machinery. Consequently, strategies which target these mechanisms represent potential targets for broad spectrum host oriented therapeutics. An important cellular signal implicated previously in EBOV budding is calcium. Indeed, host cell calcium signals are increasingly being recognized to play a role in steps of entry, replication, and transmission for a range of viruses, but if and how filoviruses and arenaviruses mobilize calcium and the precise stage of virus transmission regulated by calcium have not been defined. Here we demonstrate that expression of matrix proteins from both filoviruses and arenaviruses triggers an increase in host cytoplasmic Ca2+ concentration by a mechanism that requires host Orai1 channels. Furthermore, we demonstrate that Orai1 regulates both VLP and infectious filovirus and arenavirus production and spread. Notably, suppression of the protein that triggers Orai activation (Stromal Interaction Molecule 1, STIM1) and genetic inactivation or pharmacological blockade of Orai1 channels inhibits VLP and infectious virus egress. These findings are highly significant as they expand our understanding of host mechanisms that may broadly control enveloped RNA virus budding, and they establish Orai and STIM1 as novel targets for broad-spectrum host-oriented therapeutics to combat these emerging BSL-4 pathogens and potentially other enveloped RNA viruses that bud via similar mechanisms.


Assuntos
Arenavirus/fisiologia , Filoviridae/fisiologia , Liberação de Vírus , Animais , Cálcio/metabolismo , Canais de Cálcio/fisiologia , Células HEK293 , Células HeLa , Humanos , Proteína ORAI1 , Células Vero , Proteínas da Matriz Viral/fisiologia , Vírion/fisiologia
6.
PLoS Pathog ; 11(5): e1004900, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25993603

RESUMO

Arenaviruses are one of the largest families of human hemorrhagic fever viruses and are known to infect both mammals and snakes. Arenaviruses package a large (L) and small (S) genome segment in their virions. For segmented RNA viruses like these, novel genotypes can be generated through mutation, recombination, and reassortment. Although it is believed that an ancient recombination event led to the emergence of a new lineage of mammalian arenaviruses, neither recombination nor reassortment has been definitively documented in natural arenavirus infections. Here, we used metagenomic sequencing to survey the viral diversity present in captive arenavirus-infected snakes. From 48 infected animals, we determined the complete or near complete sequence of 210 genome segments that grouped into 23 L and 11 S genotypes. The majority of snakes were multiply infected, with up to 4 distinct S and 11 distinct L segment genotypes in individual animals. This S/L imbalance was typical: in all cases intrahost L segment genotypes outnumbered S genotypes, and a particular S segment genotype dominated in individual animals and at a population level. We corroborated sequencing results by qRT-PCR and virus isolation, and isolates replicated as ensembles in culture. Numerous instances of recombination and reassortment were detected, including recombinant segments with unusual organizations featuring 2 intergenic regions and superfluous content, which were capable of stable replication and transmission despite their atypical structures. Overall, this represents intrahost diversity of an extent and form that goes well beyond what has been observed for arenaviruses or for viruses in general. This diversity can be plausibly attributed to the captive intermingling of sub-clinically infected wild-caught snakes. Thus, beyond providing a unique opportunity to study arenavirus evolution and adaptation, these findings allow the investigation of unintended anthropogenic impacts on viral ecology, diversity, and disease potential.


Assuntos
Infecções por Arenaviridae/veterinária , Arenavirus/genética , Transmissão de Doença Infecciosa/veterinária , Rearranjo Gênico , Recombinação Genética , Serpentes/virologia , Animais , Animais de Zoológico/sangue , Animais de Zoológico/metabolismo , Animais de Zoológico/virologia , Infecções por Arenaviridae/metabolismo , Infecções por Arenaviridae/patologia , Infecções por Arenaviridae/virologia , Arenavirus/isolamento & purificação , Arenavirus/fisiologia , Sequência de Bases , Boidae/virologia , Células Cultivadas , Genoma Viral , Fígado/metabolismo , Fígado/patologia , Fígado/virologia , Dados de Sequência Molecular , Animais de Estimação/sangue , Animais de Estimação/metabolismo , Animais de Estimação/virologia , Filogenia , RNA Viral/sangue , RNA Viral/química , RNA Viral/metabolismo , Serpentes/sangue , Serpentes/metabolismo , Estados Unidos , Replicação Viral
7.
Curr Top Microbiol Immunol ; 392: 231-76, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26472215

RESUMO

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.


Assuntos
Infecções por Arenaviridae/virologia , Arenavirus/genética , Evolução Molecular , Animais , Antivirais/farmacologia , Infecções por Arenaviridae/tratamento farmacológico , Arenavirus/classificação , Arenavirus/efeitos dos fármacos , Arenavirus/fisiologia , Variação Genética , Genoma Viral , Humanos , Filogenia , Replicação Viral
8.
J Virol ; 89(16): 8428-43, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26041296

RESUMO

UNLABELLED: Certain members of the Arenaviridae family are category A agents capable of causing severe hemorrhagic fevers in humans. Specific antiviral treatments do not exist, and the only commonly used drug, ribavirin, has limited efficacy and can cause severe side effects. The discovery and development of new antivirals are inhibited by the biohazardous nature of the viruses, making them a relatively poorly understood group of human pathogens. We therefore adapted a reverse-genetics minigenome (MG) rescue system based on Junin virus, the causative agent of Argentine hemorrhagic fever, for high-throughput screening (HTS). The MG rescue system recapitulates all stages of the virus life cycle and enables screening of small-molecule libraries under biosafety containment level 2 (BSL2) conditions. The HTS resulted in the identification of four candidate compounds with potent activity against a broad panel of arenaviruses, three of which were completely novel. The target for all 4 compounds was the stage of viral entry, which positions the compounds as potentially important leads for future development. IMPORTANCE: The arenavirus family includes several members that are highly pathogenic, causing acute viral hemorrhagic fevers with high mortality rates. No specific effective treatments exist, and although a vaccine is available for Junin virus, the causative agent of Argentine hemorrhagic fever, it is licensed for use only in areas where Argentine hemorrhagic fever is endemic. For these reasons, it is important to identify specific compounds that could be developed as antivirals against these deadly viruses.


Assuntos
Antivirais/farmacologia , Infecções por Arenaviridae/prevenção & controle , Arenavirus/fisiologia , Avaliação Pré-Clínica de Medicamentos/métodos , Ensaios de Triagem em Larga Escala/métodos , Internalização do Vírus/efeitos dos fármacos , Antivirais/isolamento & purificação , Humanos , Vírus Junin/genética , Genética Reversa/métodos
9.
J Virol ; 88(1): 643-54, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24173224

RESUMO

Several arenaviruses, chiefly Lassa virus (LASV), cause hemorrhagic fever (HF) disease in humans and pose a great public health concern in the regions in which they are endemic. Moreover, evidence indicates that the worldwide-distributed prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) is a neglected human pathogen of clinical significance. The limited existing armamentarium to combat human-pathogenic arenaviruses underscores the importance of developing novel antiarenaviral drugs, a task that would be facilitated by the identification and characterization of virus-host cell factor interactions that contribute to the arenavirus life cycle. A genome-wide small interfering RNA (siRNA) screen identified sodium hydrogen exchanger 3 (NHE3) as required for efficient multiplication of LCMV in HeLa cells, but the mechanisms by which NHE activity contributed to the life cycle of LCMV remain unknown. Here we show that treatment with the NHE inhibitor 5-(N-ethyl-N-isopropyl) amiloride (EIPA) resulted in a robust inhibition of LCMV multiplication in both rodent (BHK-21) and human (A549) cells. EIPA-mediated inhibition was due not to interference with virus RNA replication, gene expression, or budding but rather to a blockade of virus cell entry. EIPA also inhibited cell entry mediated by the glycoproteins of the HF arenaviruses LASV and Junin virus (JUNV). Pharmacological and genetic studies revealed that cell entry of LCMV in A549 cells depended on actin remodeling and Pak1, suggesting a macropinocytosis-like cell entry pathway. Finally, zoniporide, an NHE inhibitor being explored as a therapeutic agent to treat myocardial infarction, inhibited LCMV propagation in culture cells. Our findings indicate that targeting NHEs could be a novel strategy to combat human-pathogenic arenaviruses.


Assuntos
Arenavirus/fisiologia , Fusão de Membrana/fisiologia , Trocadores de Sódio-Hidrogênio/fisiologia , Animais , Western Blotting , Linhagem Celular , Humanos
10.
J Virol ; 88(2): 878-89, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24198417

RESUMO

Arenaviruses merit significant interest as important human pathogens, since several of them cause severe hemorrhagic fever disease that is associated with high morbidity and significant mortality. Currently, there are no FDA-licensed arenavirus vaccines available, and current antiarenaviral therapy is limited to an off-labeled use of the nucleoside analog ribavirin, which has limited prophylactic efficacy. The pyrimidine biosynthesis inhibitor A3, which was identified in a high-throughput screen for compounds that blocked influenza virus replication, exhibits a broad-spectrum antiviral activity against negative- and positive-sense RNA viruses, retroviruses, and DNA viruses. In this study, we evaluated the antiviral activity of A3 against representative Old World (lymphocytic choriomeningitis virus) and New World (Junin virus) arenaviruses in rodent, monkey, and human cell lines. We show that A3 is significantly more efficient than ribavirin in controlling arenavirus multiplication and that the A3 inhibitory effect is in part due to its ability to interfere with viral RNA replication and transcription. We document an additive antiarenavirus effect of A3 and ribavirin, supporting the potential combination therapy of ribavirin and pyrimidine biosynthesis inhibitors for the treatment of arenavirus infections.


Assuntos
Antivirais/farmacologia , Infecções por Arenaviridae/virologia , Arenavirus/efeitos dos fármacos , Pirimidinas/antagonistas & inibidores , Animais , Infecções por Arenaviridae/metabolismo , Arenavirus/genética , Arenavirus/fisiologia , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Humanos , Pirimidinas/biossíntese , Replicação Viral/efeitos dos fármacos
11.
J Virol ; 87(7): 4071-4, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23325690

RESUMO

A recent publication indicated that overexpression of Axl, a cellular receptor that negatively regulates Toll-like receptor signaling, enhanced the entry of viruses pseudotyped with the glycoprotein of lymphocytic choriomeningitis virus (LCMV) in vitro. In testing the biological relevance of these observations, we found differences in neither viral kinetics between LCMV infections of Axl(-/-) and wild-type mice nor T-cell responses prior to spontaneous viral clearance. Thus, Axl is not required for productive LCMV infection of mice.


Assuntos
Infecções por Arenaviridae/fisiopatologia , Arenavirus/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Internalização do Vírus , Animais , Arenavirus/fisiologia , Fluorescência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Receptor Tirosina Quinase Axl
12.
J Virol ; 86(19): 10759-65, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22837210

RESUMO

Arenaviruses are rodent-borne viruses with a bisegmented RNA genome. A genetically unique arenavirus, Lujo virus, was recently discovered as the causal agent of a nosocomial outbreak of acute febrile illness with hemorrhagic manifestations in Zambia and South Africa. The outbreak had a case fatality rate of 80%. A reverse genetics system to rescue infectious Lujo virus from cDNA was established to investigate the biological properties of this virus. Sequencing the genomic termini showed unique nucleotides at the 3' terminus of the S segment promoter element. While developing this system, we discovered that reconstructing infectious Lujo virus using the previously reported L segment intergenic region (IGR), comprising the arenaviral transcription termination signal, yielded an attenuated Lujo virus. Resequencing revealed that the correct L segment IGR was 36 nucleotides longer, and incorporating it into the reconstructed Lujo virus restored the growth rate to that of the authentic clinical virus isolate. These additional nucleotides were predicted to more than double the free energy of the IGR main stem-loop structure. In addition, incorporating the newly determined L-IGR into a replicon reporter system enhanced the expression of a luciferase reporter L segment. Overall, these results imply that an extremely stable secondary structure within the L-IGR is critical for Lujo virus propagation and viral protein production. The technology for producing recombinant Lujo virus now provides a method to precisely investigate the molecular determinants of virulence of this newly identified pathogen.


Assuntos
Arenavirus/genética , RNA Viral/genética , Regiões 3' não Traduzidas , Animais , Arenavirus/fisiologia , Sequência de Bases , Cricetinae , DNA Complementar/metabolismo , Genes Reporter , Modelos Genéticos , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Nucleotídeos/genética , Plasmídeos/metabolismo , Vírus de RNA/genética , África do Sul , Fatores de Tempo , Virulência , Zâmbia
13.
J Virol ; 86(11): 6138-45, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22438561

RESUMO

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.


Assuntos
Arenavirus/fisiologia , Glicoproteínas/metabolismo , Sinais Direcionadores de Proteínas , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Sequência de Aminoácidos , Animais , Arenavirus/genética , Chlorocebus aethiops , Glicoproteínas/genética , Modelos Moleculares , Dados de Sequência Molecular , Células Vero , Proteínas do Envelope Viral/genética
14.
J Virol ; 86(18): 9794-801, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22761375

RESUMO

Arenaviruses can cause severe hemorrhagic fever diseases in humans, with limited prophylactic or therapeutic measures. A small RING-domain viral protein Z has been shown to mediate the formation of virus-like particles and to inhibit viral RNA synthesis, although its biological roles in an infectious viral life cycle have not been directly addressed. By taking advantage of the available reverse genetics system for a model arenavirus, Pichinde virus (PICV), we provide the direct evidence for the essential biological roles of the Z protein's conserved residues, including the G2 myristylation site, the conserved C and H residues of RING domain, and the poorly characterized C-terminal L79 and P80 residues. Dicodon substitutions within the late (L) domain (PSAPPYEP) of the PICV Z protein, although producing viable mutant viruses, have significantly reduced virus growth, a finding suggestive of an important role for the intact L domain in viral replication. Further structure-function analyses of both PICV and Lassa fever virus Z proteins suggest that arenavirus Z proteins have similar molecular mechanisms in mediating their multiple functions, with some interesting variations, such as the role of the G2 residue in blocking viral RNA synthesis. In summary, our studies have characterized the biological roles of the Z protein in an infectious arenavirus system and have shed important light on the distinct functions of its domains in virus budding and viral RNA regulation, the knowledge of which may lead to the development of novel antiviral drugs.


Assuntos
Arenavirus/fisiologia , Proteínas Virais/fisiologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Infecções por Arenaviridae/etiologia , Infecções por Arenaviridae/virologia , Arenavirus/genética , Arenavirus/patogenicidade , Linhagem Celular , Sequência Conservada , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Vírus Pichinde/genética , Vírus Pichinde/patogenicidade , Vírus Pichinde/fisiologia , Estrutura Terciária de Proteína , RNA Viral/biossíntese , Homologia de Sequência de Aminoácidos , Proteínas Virais/química , Proteínas Virais/genética , Liberação de Vírus/genética , Liberação de Vírus/fisiologia , Replicação Viral/genética , Replicação Viral/fisiologia
15.
Viruses ; 14(9)2022 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-36146793

RESUMO

Mastomys natalensis is the natural host of various arenaviruses, including the human-pathogenic Lassa virus. Homologous arenaviruses, defined here as those having M. natalensis as a natural host, can establish long-lasting infection in M. natalensis, while these animals rapidly clear arenaviruses having another rodent species as a natural host (heterologous viruses). Little is known about the mechanisms behind the underlying arenavirus-host barriers. The innate immune system, particularly the type I interferon (IFN) response, might play a role. In this study, we developed and validated RT-PCR assays to analyse the expression of M. natalensis interferon-stimulated genes (ISGs). We then used these assays to study if homologous and heterologous viruses induce different IFN responses in M. natalensis cells. Infection experiments were performed with the homologous Lassa and Morogoro viruses and the related but heterologous Mobala virus. Compared to the direct induction with IFN or Poly(I:C), arenaviruses generally induced a weak IFN response. However, the ISG-expression profiles of homologous and heterologous viruses were similar. Our data indicate that, at least in M. natalensis cells, the IFN system is not a major factor in the virus-host barrier for arenaviruses. Our system provides a valuable tool for future in vivo investigation of arenavirus host restrictions at the level of the innate immune response.


Assuntos
Infecções por Arenaviridae , Arenavirus , Interferon Tipo I , Animais , Arenavirus/fisiologia , Humanos , Imunidade Inata , Murinae , Tanzânia
16.
Viruses ; 13(5)2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-34067011

RESUMO

Natural hosts of most arenaviruses are rodents. The human-pathogenic Lassa virus and several non-pathogenic arenaviruses such as Morogoro virus (MORV) share the same host species, namely Mastomys natalensis (M. natalensis). In this study, we investigated the history of infection and virus transmission within the natural host population. To this end, we infected M. natalensis at different ages with MORV and measured the health status of the animals, virus load in blood and organs, the development of virus-specific antibodies, and the ability of the infected individuals to transmit the virus. To explore the impact of the lack of evolutionary virus-host adaptation, experiments were also conducted with Mobala virus (MOBV), which does not share M. natalensis as a natural host. Animals infected with MORV up to two weeks after birth developed persistent infection, seroconverted and were able to transmit the virus horizontally. Animals older than two weeks at the time of infection rapidly cleared the virus. In contrast, MOBV-infected neonates neither developed persistent infection nor were able to transmit the virus. In conclusion, we demonstrate that MORV is able to develop persistent infection in its natural host, but only after inoculation shortly after birth. A related arenavirus that is not evolutionarily adapted to M. natalensis is not able to establish persistent infection. Persistently infected animals appear to be important to maintain virus transmission within the host population.


Assuntos
Infecções por Arenaviridae/veterinária , Arenavirus/fisiologia , Reservatórios de Doenças/virologia , Murinae/virologia , Animais , Animais Recém-Nascidos , Arenavirus/classificação , Especificidade de Hospedeiro , Doenças dos Roedores/virologia , Replicação Viral
17.
Viruses ; 13(6)2021 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-34073735

RESUMO

Several of the human-pathogenic arenaviruses cause hemorrhagic fever and have to be handled under biosafety level 4 conditions, including Lassa virus. Rapid and safe inactivation of specimens containing these viruses is fundamental to enable downstream processing for diagnostics or research under lower biosafety conditions. We established a protocol to test the efficacy of inactivation methods using the low-pathogenic Morogoro arenavirus as surrogate for the related highly pathogenic viruses. As the validation of chemical inactivation methods in cell culture systems is difficult due to cell toxicity of commonly used chemicals, we employed filter devices to remove the chemical and concentrate the virus after inactivation and before inoculation into cell culture. Viral replication in the cells was monitored over 4 weeks by using indirect immunofluorescence and immunofocus assay. The performance of the protocol was verified using published inactivation methods including chemicals and heat. Ten additional methods to inactivate virus in infected cells or cell culture supernatant were validated and shown to reduce virus titers to undetectable levels. In summary, we provide a robust protocol for the validation of chemical and physical inactivation of arenaviruses in cell culture, which can be readily adapted to different inactivation methods and specimen matrices.


Assuntos
Arenavirus/fisiologia , Desinfecção/métodos , Inativação de Vírus , Animais , Técnicas de Cultura de Células , Linhagem Celular , Células Cultivadas , Chlorocebus aethiops , Desinfecção/normas , Humanos , Reprodutibilidade dos Testes , Manejo de Espécimes/métodos , Células Vero
18.
Viruses ; 13(7)2021 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-34203149

RESUMO

Lujo virus (LUJV), a highly pathogenic arenavirus, was first identified in 2008 in Zambia. To aid the identification of effective therapeutics for LUJV, we developed a recombinant reporter virus system, confirming reporter LUJV comparability with wild-type virus and its utility in high-throughput antiviral screening assays. Using this system, we evaluated compounds with known and unknown efficacy against related arenaviruses, with the aim of identifying LUJV-specific and potential new pan-arenavirus antivirals. We identified six compounds demonstrating robust anti-LUJV activity, including several compounds with previously reported activity against other arenaviruses. These data provide critical evidence for developing broad-spectrum antivirals against high-consequence arenaviruses.


Assuntos
Antivirais/farmacologia , Arenavirus/efeitos dos fármacos , Lujo virus/efeitos dos fármacos , Animais , Infecções por Arenaviridae/tratamento farmacológico , Infecções por Arenaviridae/virologia , Arenavirus/fisiologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Genoma Viral , Proteínas de Fluorescência Verde/genética , Humanos , Lujo virus/genética , Lujo virus/fisiologia , Testes de Sensibilidade Microbiana , Proteínas Recombinantes , Células Vero , Internalização do Vírus/efeitos dos fármacos
19.
J Exp Clin Cancer Res ; 39(1): 34, 2020 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-32041643

RESUMO

Odontogenic tumors (OT) are considered rare events and their epidemiologic data are scarce and under-estimated in developing countries because there is no systematic collection of clinical features including histological analyses of the tissue samples. Furthermore, there is an underestimation of the disease relevance and affected people are often marginalized in spite of severe functional impairment of aero-digestive tract. Etiology of OT in humans is still unknown and it represents an important therapeutic and diagnostic challenge.Lassa fever is an acute viral haemorrhagic illness caused by Lassa virus, a member of the arenavirus family of viruses. The disease is endemic in the rodent population in West-East Africa. Humans usually become infected with Lassa virus through exposure to the food or household items contaminated with urine or feces of infected rats. It is also reported person-to-person infections. About 80% of people infected by Lassa virus have no symptoms but the virus establishes a life-long persistent infection.The present commentary significance is to start, for the first time ever, a systematic collection of clinical features and tissue sample collection at the St. Mary's Hospital in Lacor (Gulu) North Uganda where the considered pathologies have an important frequency. The systematic collection will allow to corroborate the possible association between arenaviruses infection and pathogenesis of odontogenic tumors in humans.


Assuntos
Infecções por Arenaviridae/complicações , Infecções por Arenaviridae/virologia , Arenavirus/fisiologia , Transformação Celular Viral , Tumores Odontogênicos/etiologia , Infecções por Arenaviridae/epidemiologia , Biópsia , Suscetibilidade a Doenças , Humanos , Febre Lassa/complicações , Febre Lassa/virologia , Vírus Lassa , Tumores Odontogênicos/diagnóstico , Tumores Odontogênicos/epidemiologia , Uganda
20.
J Virol ; 82(21): 10932-9, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18768973

RESUMO

The arenavirus envelope glycoprotein (GPC) mediates viral entry through pH-induced membrane fusion in the endosome. This crucial process in the viral life cycle can be specifically inhibited in the New World arenaviruses by the small-molecule compound ST-294. Here, we show that ST-294 interferes with GPC-mediated membrane fusion by targeting the interaction of the G2 fusion subunit with the stable signal peptide (SSP). We demonstrate that amino acid substitutions at lysine-33 of the Junín virus SSP confer resistance to ST-294 and engender de novo sensitivity to ST-161, a chemically distinct inhibitor of the Old World Lassa fever virus. These compounds, as well as a broadly active inhibitor, ST-193, likely share a molecular target at the SSP-G2 interface. We also show that both ST-294 and ST-193 inhibit pH-induced dissociation of the G1 receptor-binding subunit from GPC, a process concomitant with fusion activation. Interestingly, the inhibitory activity of these molecules can in some cases be overcome by further lowering the pH used for activation. Our results suggest that these small molecules act to stabilize the prefusion GPC complex against acidic pH. The pH-sensitive interaction between SSP and G2 in GPC represents a robust molecular target for the development of antiviral compounds for the treatment of arenavirus hemorrhagic fevers.


Assuntos
Arenavirus/efeitos dos fármacos , Arenavirus/fisiologia , Fusão de Membrana/efeitos dos fármacos , Fusão de Membrana/fisiologia , Sulfonamidas/farmacologia , Ureia/análogos & derivados , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus/efeitos dos fármacos , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Antivirais/farmacologia , Farmacorresistência Viral , Concentração de Íons de Hidrogênio , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Alinhamento de Sequência , Ureia/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA