Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Viruses ; 13(7)2021 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-34203149

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Arenavirus/efectos de los fármacos , Lujo virus/efectos de los fármacos , Animales , Infecciones por Arenaviridae/tratamiento farmacológico , Infecciones por Arenaviridae/virología , Arenavirus/fisiología , Línea Celular Tumoral , Chlorocebus aethiops , Genoma Viral , Proteínas Fluorescentes Verdes/genética , Humanos , Lujo virus/genética , Lujo virus/fisiología , Pruebas de Sensibilidad Microbiana , Proteínas Recombinantes , Células Vero , Internalización del Virus/efectos de los fármacos
2.
J Exp Clin Cancer Res ; 39(1): 34, 2020 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-32041643

RESUMEN

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.


Asunto(s)
Infecciones por Arenaviridae/complicaciones , Infecciones por Arenaviridae/virología , Arenavirus/fisiología , Transformación Celular Viral , Tumores Odontogénicos/etiología , Infecciones por Arenaviridae/epidemiología , Biopsia , Susceptibilidad a Enfermedades , Humanos , Fiebre de Lassa/complicaciones , Fiebre de Lassa/virología , Virus Lassa , Tumores Odontogénicos/diagnóstico , Tumores Odontogénicos/epidemiología , Uganda
3.
Viruses ; 11(3)2019 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-30909570

RESUMEN

Lassa virus (LASV) and Mopeia virus (MOPV) are two closely related Old-World mammarenaviruses. LASV causes severe hemorrhagic fever with high mortality in humans, whereas no case of MOPV infection has been reported. Comparing MOPV and LASV is a powerful strategy to unravel pathogenic mechanisms that occur during the course of pathogenic arenavirus infection. We used a yeast two-hybrid approach to identify cell partners of MOPV and LASV Z matrix protein in which two autophagy adaptors were identified, NDP52 and TAX1BP1. Autophagy has emerged as an important cellular defense mechanism against viral infections but its role during arenavirus infection has not been shown. Here, we demonstrate that autophagy is transiently induced by MOPV, but not LASV, in infected cells two days after infection. Impairment of the early steps of autophagy significantly decreased the production of MOPV and LASV infectious particles, whereas a blockade of the degradative steps impaired only MOPV infectious particle production. Our study provides insights into the role played by autophagy during MOPV and LASV infection and suggests that this process could partially explain their different pathogenicity.


Asunto(s)
Arenavirus/fisiología , Autofagia , Virus Lassa/fisiología , Animales , Arenavirus/patogenicidad , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Virus Lassa/patogenicidad , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Técnicas del Sistema de Dos Híbridos , Células Vero
4.
Nat Commun ; 8: 14447, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28248314

RESUMEN

Immune-mediated effector molecules can limit cancer growth, but lack of sustained immune activation in the tumour microenvironment restricts antitumour immunity. New therapeutic approaches that induce a strong and prolonged immune activation would represent a major immunotherapeutic advance. Here we show that the arenaviruses lymphocytic choriomeningitis virus (LCMV) and the clinically used Junin virus vaccine (Candid#1) preferentially replicate in tumour cells in a variety of murine and human cancer models. Viral replication leads to prolonged local immune activation, rapid regression of localized and metastatic cancers, and long-term disease control. Mechanistically, LCMV induces antitumour immunity, which depends on the recruitment of interferon-producing Ly6C+ monocytes and additionally enhances tumour-specific CD8+ T cells. In comparison with other clinically evaluated oncolytic viruses and to PD-1 blockade, LCMV treatment shows promising antitumoural benefits. In conclusion, therapeutically administered arenavirus replicates in cancer cells and induces tumour regression by enhancing local immune responses.


Asunto(s)
Arenavirus/fisiología , Vigilancia Inmunológica , Interferón Tipo I/metabolismo , Neoplasias/inmunología , Neoplasias/virología , Replicación Viral/fisiología , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Humanos , Activación de Linfocitos/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Ratones Endogámicos C57BL , Monocitos/metabolismo , Neoplasias/irrigación sanguínea , Virus Oncolíticos/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo
5.
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
6.
J Virol ; 87(7): 4071-4, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23325690

RESUMEN

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.


Asunto(s)
Infecciones por Arenaviridae/fisiopatología , Arenavirus/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Internalización del Virus , Animales , Arenavirus/fisiología , Fluorescencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Tirosina Quinasa del Receptor Axl
7.
Viruses ; 4(11): 2973-3011, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-23202512

RESUMEN

Arenaviruses are a family of enveloped negative-stranded RNA viruses that can cause severe human disease ranging from encephalitis symptoms to fulminant hemorrhagic fever. The bi­segmented RNA genome encodes four polypeptides: the nucleoprotein NP, the surface glycoprotein GP, the polymerase L, and the RING finger protein Z. Although it is the smallest arenavirus protein with a length of 90 to 99 amino acids and a molecular weight of approx. 11 kDa, the Z protein has multiple functions in the viral life cycle including (i) regulation of viral RNA synthesis, (ii) orchestration of viral assembly and budding, (iii) interaction with host cell proteins, and (iv) interferon antagonism. In this review, we summarize our current understanding of the structural and functional role of the Z protein in the arenavirus replication cycle.


Asunto(s)
Arenavirus/fisiología , Proteínas Virales/metabolismo , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Infecciones por Arenaviridae/tratamiento farmacológico , Infecciones por Arenaviridae/virología , Genoma Viral , Humanos , Dominios RING Finger , Interferencia de ARN , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/química , Proteínas Virales/genética , Virión/ultraestructura , Replicación Viral
8.
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
9.
J Gen Virol ; 89(Pt 11): 2713-2722, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18931067

RESUMEN

alpha-Dystroglycan (alpha-DG) is a ubiquitously expressed molecule that has been identified as a cellular receptor for lymphocytic choriomeningitis virus (LCMV) and other arenaviruses. Recently, it was demonstrated that LCMV receptor function is critically dependent on post-translational modifications, namely glycosylation. In particular, it was shown that O-mannosylation, a rare type of mammalian O-linked glycosylation, is important in determining the binding of LCMV to its cellular receptor. All studies carried out so far showed a dependence on glycosylation in LCMV receptor function in vitro. This work extended these studies to two in vivo models of alpha-DG hypoglycosylation. The results confirm earlier findings on the in vitro dependence of carbohydrate modifications in LCMV receptor function. However, experiments in animal models showed that this dependence was only very weak in vivo. It is likely that alternative receptors or alternative entry pathways may account for this attenuated in vivo phenotype.


Asunto(s)
Distroglicanos/metabolismo , Virus de la Coriomeningitis Linfocítica/fisiología , Procesamiento Proteico-Postraduccional , Receptores Virales/metabolismo , Animales , Animales Recién Nacidos , Arenavirus/fisiología , Astrocitos/fisiología , Astrocitos/virología , Células de la Médula Ósea/fisiología , Células de la Médula Ósea/virología , Células Dendríticas/fisiología , Células Dendríticas/virología , Citometría de Flujo , Leucocitos/fisiología , Leucocitos/virología , Macrófagos Peritoneales/fisiología , Macrófagos Peritoneales/virología , Ratones , Ratones Endogámicos C57BL , Modelos Animales
10.
J Virol ; 82(21): 10932-9, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18768973

RESUMEN

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.


Asunto(s)
Arenavirus/efectos de los fármacos , Arenavirus/fisiología , Fusión de Membrana/efectos de los fármacos , Fusión de Membrana/fisiología , Sulfonamidas/farmacología , Urea/análogos & derivados , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus/efectos de los fármacos , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Antivirales/farmacología , Farmacorresistencia Viral , Concentración de Iones de Hidrógeno , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Alineación de Secuencia , Urea/farmacología
11.
J Virol ; 81(17): 9451-60, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17581989

RESUMEN

Generation of infectious arenavirus-like particles requires the virus RING finger Z protein and surface glycoprotein precursor (GPC) and the correct processing of GPC into GP1, GP2, and a stable signal peptide (SSP). Z is the driving force of arenavirus budding, whereas the GP complex (GPc), consisting of hetero-oligomers of SSP, GP1, and GP2, forms the viral envelope spikes that mediate receptor recognition and cell entry. Based on the roles played by Z and GP in the arenavirus life cycle, we hypothesized that Z and the GPc should interact in a manner required for virion formation. Here, using confocal microscopy and coimmunoprecipitation assays, we provide evidence for subcellular colocalization and biochemical interaction, respectively, of Z and the GPc. Our results from mutation-function analysis reveal that Z myristoylation, but not the Z late (L) or RING domain, is required for Z-GPc interaction. Moreover, Z interacted directly with SSP in the absence of other components of the GPc. We obtained similar results with Z and GPC from the prototypical arenavirus lymphocytic choriomeningitis virus and the hemorrhagic fever arenavirus Lassa fever virus.


Asunto(s)
Arenavirus/fisiología , Procesamiento Proteico-Postraduccional , Proteínas Virales/metabolismo , Ensamble de Virus/fisiología , Animales , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Línea Celular , Citoplasma/química , Glicoproteínas/química , Glicoproteínas/metabolismo , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular , Microscopía Confocal , Unión Proteica , Proteínas de Unión al ARN , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Proteínas de la Matriz Viral/metabolismo , Proteínas Virales/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA