Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Virol ; 98(3): e0188323, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38376197

RESUMEN

Many viruses, including mammarenaviruses, have evolved mechanisms to counteract different components of the host cell innate immunity, which is required to facilitate robust virus multiplication. The double-stranded RNA (dsRNA) sensor protein kinase receptor (PKR) pathway plays a critical role in the cell anti-viral response. Whether PKR can restrict the multiplication of the Old World mammarenavirus lymphocytic choriomeningitis virus (LCMV) and the mechanisms by which LCMV may counteract the anti-viral functions of PKR have not yet been investigated. Here we present evidence that LCMV infection results in very limited levels of PKR activation, but LCMV multiplication is enhanced in the absence of PKR. In contrast, infection with a recombinant LCMV with a mutation affecting the 3'-5' exonuclease (ExoN) activity of the viral nucleoprotein resulted in robust PKR activation in the absence of detectable levels of dsRNA, which was associated with severely restricted virus multiplication that was alleviated in the absence of PKR. However, pharmacological inhibition of PKR activation resulted in reduced levels of LCMV multiplication. These findings uncovered a complex role of the PKR pathway in LCMV-infected cells involving both pro- and anti-viral activities.IMPORTANCEAs with many other viruses, the prototypic Old World mammarenavirus LCMV can interfere with the host cell innate immune response to infection, which includes the dsRNA sensor PKR pathway. A detailed understanding of LCMV-PKR interactions can provide novel insights about mammarenavirus-host cell interactions and facilitate the development of effective anti-viral strategies against human pathogenic mammarenaviruses. In the present work, we present evidence that LCMV multiplication is enhanced in PKR-deficient cells, but pharmacological inhibition of PKR activation unexpectedly resulted in severely restricted propagation of LCMV. Likewise, we document a robust PKR activation in LCMV-infected cells in the absence of detectable levels of dsRNA. Our findings have revealed a complex role of the PKR pathway during LCMV infection and uncovered the activation of PKR as a druggable target for the development of anti-viral drugs against human pathogenic mammarenaviruses.


Asunto(s)
Arenaviridae , Coriomeningitis Linfocítica , Humanos , Arenaviridae/metabolismo , Línea Celular , Proteínas Quinasas/metabolismo , Interacciones Huésped-Patógeno , Virus de la Coriomeningitis Linfocítica/metabolismo , Proteínas Portadoras , Antivirales , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
2.
Curr Opin Struct Biol ; 79: 102561, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36857816

RESUMEN

Viruses are obligatory parasites that can replicate only inside host cells. Therefore, the evolutionary drive to enter cells is immense, leading to diversification in the cell-entry strategies of viruses. One of the most critical steps for cell entry is the recognition of the target cell, a process driven by the formation of viral/host macromolecular complexes. The accumulation of recent structural data for viruses within the arenaviridae family allows us to examine how different viral species from the same viral family utilize evolutionarily-related viral glycoproteins to engage with a variety of different cellular receptors. These structural data, compared to other viruses from the coronaviridae family, hint about possible routes that such viruses use for evolving new receptor-binding capabilities, allowing them to switch from one receptor to another.


Asunto(s)
Arenaviridae , Virus , Arenaviridae/metabolismo , Virus/metabolismo , Receptores de Superficie Celular/metabolismo , Unión Proteica , Sustancias Macromoleculares
3.
Curr Top Microbiol Immunol ; 439: 265-303, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36592249

RESUMEN

Members of the family Arenaviridae are classified into four genera: Antennavirus, Hartmanivirus, Mammarenavirus, and Reptarenavirus. Reptarenaviruses and hartmaniviruses infect (captive) snakes and have been shown to cause boid inclusion body disease (BIBD). Antennaviruses have genomes consisting of 3, rather than 2, segments, and were discovered in actinopterygian fish by next-generation sequencing but no biological isolate has been reported yet. The hosts of mammarenaviruses are mainly rodents and infections are generally asymptomatic. Current knowledge about the biology of reptarenaviruses, hartmaniviruses, and antennaviruses is very limited and their zoonotic potential is unknown. In contrast, some mammarenaviruses are associated with zoonotic events that pose a threat to human health. This review will focus on mammarenavirus genetic diversity and its biological implications. Some mammarenaviruses including lymphocytic choriomeningitis virus (LCMV) are excellent experimental model systems for the investigation of acute and persistent viral infections, whereas others including Lassa (LASV) and Junin (JUNV) viruses, the causative agents of Lassa fever (LF) and Argentine hemorrhagic fever (AHF), respectively, are important human pathogens. Mammarenaviruses were thought to have high degree of intra-and inter-species amino acid sequence identities, but recent evidence has revealed a high degree of mammarenavirus genetic diversity in the field. Moreover, closely related mammarenavirus can display dramatic phenotypic differences in vivo. These findings support a role of genetic variability in mammarenavirus adaptability and pathogenesis. Here, we will review the molecular biology of mammarenaviruses, phylogeny, and evolution, as well as the quasispecies dynamics of mammarenavirus populations and their biological implications.


Asunto(s)
Arenaviridae , Animales , Humanos , Arenaviridae/genética , Arenaviridae/metabolismo , Roedores , Variación Genética
4.
Viruses ; 14(7)2022 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-35891543

RESUMEN

Replication-competent reporter-expressing viruses are crucial tools in molecular virology with applications that range from antiviral screening to live-cell imaging of protein spatiotemporal dynamics. However, there is currently little information available regarding viable strategies to develop reporter-expressing arenaviruses. To address this, we used Tacaribe virus (TCRV), an apathogenic BSL2 arenavirus, to assess the feasibility of different reporter expression approaches. We first generated trisegmented TCRV viruses with either the glycoprotein (GP) or nucleoprotein (NP) replaced by a reporter (GFP, mCherry, or nanoluciferase). These viruses were all viable, but showed marked differences in brightness and attenuation. Next, we generated terminal fusions with each of the TCRV proteins (i.e., NP, GP, polymerase (L), matrix protein (Z)) either with or without a T2A self-cleavage site. We tested both the function of the reporter-fused proteins alone, and the viability of corresponding recombinant TCRVs. We successfully rescued viruses with both direct and cleavable reporter fusions at the C-terminus of Z, as well as cleavable N-terminal fusions with NP. These viruses all displayed detectable reporter activity, but were also moderately attenuated. Finally, reporter proteins were inserted into a flexible hinge region within L. These viruses were also viable and showed moderate attenuation; however, reporter expression was only detectable for the luminescent virus. These strategies provide an exciting range of new tools for research into the molecular biology of TCRV that can likely also be adapted to other arenaviruses.


Asunto(s)
Arenaviridae , Arenavirus , Arenavirus del Nuevo Mundo , Arenaviridae/genética , Arenaviridae/metabolismo , Arenavirus/genética , Arenavirus del Nuevo Mundo/genética , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , Replicación Viral
5.
Nat Commun ; 13(1): 558, 2022 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-35091550

RESUMEN

Five New World mammarenaviruses (NWMs) cause life-threatening hemorrhagic fever (HF). Cellular entry by these viruses is mediated by human transferrin receptor 1 (hTfR1). Here, we demonstrate that an antibody (ch128.1/IgG1) which binds the apical domain of hTfR1, potently inhibits infection of attenuated and pathogenic NWMs in vitro. Computational docking of the antibody Fab crystal structure onto the known structure of hTfR1 shows an overlapping receptor-binding region shared by the Fab and the viral envelope glycoprotein GP1 subunit that binds hTfR1, and we demonstrate competitive inhibition of NWM GP1 binding by ch128.1/IgG1 as the principal mechanism of action. Importantly, ch128.1/IgG1 protects hTfR1-expressing transgenic mice against lethal NWM challenge. Additionally, the antibody is well-tolerated and only partially reduces ferritin uptake. Our findings provide the basis for the development of a novel, host receptor-targeted antibody therapeutic broadly applicable to the treatment of HF of NWM etiology.


Asunto(s)
Antígenos CD/metabolismo , Arenaviridae/metabolismo , Fiebre Hemorrágica Americana/metabolismo , Receptores de Transferrina/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Células A549 , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , Antígenos CD/inmunología , Arenaviridae/efectos de los fármacos , Arenaviridae/fisiología , Chlorocebus aethiops , Fiebre Hemorrágica Americana/prevención & control , Fiebre Hemorrágica Americana/virología , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Virus Junin/efectos de los fármacos , Virus Junin/fisiología , Ratones Endogámicos C57BL , Ratones Transgénicos , Simulación del Acoplamiento Molecular , Unión Proteica/efectos de los fármacos , Receptores de Transferrina/antagonistas & inhibidores , Receptores de Transferrina/inmunología , Células Vero
6.
Sci Rep ; 11(1): 21048, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34702948

RESUMEN

Viruses need cells for their replication and, therefore, ways to hijack cellular functions. Mitochondria play fundamental roles within the cell in metabolism, immunity and regulation of homeostasis due to which some viruses aim to alter mitochondrial functions. Herein we show that the nucleoprotein (NP) of arenaviruses enters the mitochondria of infected cells, affecting the mitochondrial morphology. Reptarenaviruses cause boid inclusion body disease (BIBD) that is characterized, especially in boas, by the formation of cytoplasmic inclusion bodies (IBs) comprising reptarenavirus NP within the infected cells. We initiated this study after observing electron-dense material reminiscent of IBs within the mitochondria of reptarenavirus infected boid cell cultures in an ultrastructural study. We employed immuno-electron microscopy to confirm that the mitochondrial inclusions indeed contain reptarenavirus NP. Mutations to a putative N-terminal mitochondrial targeting signal (MTS), identified via software predictions in both mamm- and reptarenavirus NPs, did not affect the mitochondrial localization of NP, suggesting that it occurs independently of MTS. In support of MTS-independent translocation, we did not detect cleavage of the putative MTSs of arenavirus NPs in reptilian or mammalian cells. Furthermore, in vitro translated NPs could not enter isolated mitochondria, suggesting that the translocation requires cellular factors or conditions. Our findings suggest that MTS-independent mitochondrial translocation of NP is a shared feature among arenaviruses. We speculate that by targeting the mitochondria arenaviruses aim to alter mitochondrial metabolism and homeostasis or affect the cellular defense.


Asunto(s)
Arenaviridae/metabolismo , Boidae/virología , Cuerpos de Inclusión Viral/metabolismo , Mitocondrias/metabolismo , Mitocondrias/virología , Nucleoproteínas/metabolismo , Animales , Arenaviridae/clasificación , Arenaviridae/genética , Chlorocebus aethiops , Cuerpos de Inclusión Viral/genética , Mitocondrias/genética , Nucleoproteínas/genética , Células Vero
7.
Viruses ; 13(9)2021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34578344

RESUMEN

Lassa virus (LASV)-a member of the family Arenaviridae-causes Lassa fever in humans and is endemic in West Africa. Currently, no approved drugs are available. We screened 2480 small compounds for their potential antiviral activity using pseudotyped vesicular stomatitis virus harboring the LASV glycoprotein (VSV-LASVGP) and a related prototypic arenavirus, lymphocytic choriomeningitis virus (LCMV). Follow-up studies confirmed that CP100356 hydrochloride (CP100356), a specific P-glycoprotein (P-gp) inhibitor, suppressed VSV-LASVGP, LCMV, and LASV infection with half maximal inhibitory concentrations of 0.52, 0.54, and 0.062 µM, respectively, without significant cytotoxicity. Although CP100356 did not block receptor binding at the cell surface, it inhibited low-pH-dependent membrane fusion mediated by arenavirus glycoproteins. P-gp downregulation did not cause a significant reduction in either VSV-LASVGP or LCMV infection, suggesting that P-gp itself is unlikely to be involved in arenavirus entry. Finally, our data also indicate that CP100356 inhibits the infection by other mammarenaviruses. Thus, our findings suggest that CP100356 can be considered as an effective virus entry inhibitor for LASV and other highly pathogenic mammarenaviruses.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/efectos de los fármacos , Arenaviridae/metabolismo , Isoquinolinas/farmacología , Virus Lassa/efectos de los fármacos , Quinazolinas/farmacología , Internalización del Virus/efectos de los fármacos , Animales , Antivirales/farmacología , Chlorocebus aethiops , Humanos , Fiebre de Lassa/tratamiento farmacológico , Fiebre de Lassa/virología , Virus de la Coriomeningitis Linfocítica , Receptores Virales , Células Vero , Estomatitis Vesicular/virología , Inhibidores de Proteínas Virales de Fusión/farmacología
8.
Viruses ; 12(7)2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32708976

RESUMEN

Arenaviridae is a family of viruses harbouring important emerging pathogens belonging to the Bunyavirales order. Like in other segmented negative strand RNA viruses, the nucleoprotein (NP) is a major actor of the viral life cycle being both (i) the necessary co-factor of the polymerase present in the L protein, and (ii) the last line of defence of the viral genome (vRNA) by physically hiding its presence in the cytoplasm. The NP is also one of the major players interfering with the immune system. Several structural studies of NP have shown that it features two domains: a globular RNA binding domain (NP-core) in its N-terminal and an exonuclease domain (ExoN) in its C-terminal. Further studies have observed that significant conformational changes are necessary for RNA encapsidation. In this review we revisited the most recent structural and functional data available on Arenaviridae NP, compared to other Bunyavirales nucleoproteins and explored the structural and functional implications. We review the variety of structural motif extensions involved in NP-NP binding mode. We also evaluate the major functional implications of NP interactome and the role of ExoN, thus making the NP a target of choice for future vaccine and antiviral therapy.


Asunto(s)
Arenaviridae/metabolismo , Proteínas de la Nucleocápside/metabolismo , Ensamble de Virus , Arenaviridae/fisiología , Proteínas de la Nucleocápside/fisiología , Estructura Terciaria de Proteína
9.
J Virol ; 94(11)2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32238580

RESUMEN

Boid inclusion body disease (BIBD) is a transmissible viral disease of captive snakes that causes severe losses in snake collections worldwide. It is caused by reptarenavirus infection, which can persist over several years without overt signs but is generally associated with the eventual death of the affected snakes. Thus far, reports have confirmed the existence of reptarenaviruses in captive snakes in North America, Europe, Asia, and Australia, but there is no evidence that it also occurs in wild snakes. BIBD affects boa species within the subfamily Boinae and pythons in the family Pythonidae, the habitats of which do not naturally overlap. Here, we studied Brazilian captive snakes with BIBD using a metatranscriptomic approach, and we report the identification of novel reptarenaviruses, hartmaniviruses, and a new species in the family Chuviridae The reptarenavirus L segments identified are divergent enough to represent six novel species, while we found only a single novel reptarenavirus S segment. Until now, hartmaniviruses had been identified only in European captive boas with BIBD, and the present results increase the number of known hartmaniviruses from four to six. The newly identified chuvirus showed 38.4%, 40.9%, and 48.1% amino acid identity to the nucleoprotein, glycoprotein, and RNA-dependent RNA polymerase, respectively, of its closest relative, Guangdong red-banded snake chuvirus-like virus. Although we cannot rule out the possibility that the found viruses originated from imported snakes, the results suggest that the viruses could circulate in indigenous snake populations.IMPORTANCE Boid inclusion body disease (BIBD), caused by reptarenavirus infection, affects captive snake populations worldwide, but the reservoir hosts of reptarenaviruses remain unknown. Here, we report the identification of novel reptarenaviruses, hartmaniviruses, and a chuvirus in captive Brazilian boas with BIBD. Three of the four snakes studied showed coinfection with all three viruses, and one of the snakes harbored three novel reptarenavirus L segments and one novel S segment. The samples originated from collections with Brazilian indigenous snakes only, which could indicate that these viruses circulate in wild snakes. The findings could further indicate that boid snakes are the natural reservoir of reptarena- and hartmaniviruses commonly found in captive snakes. The snakes infected with the novel chuvirus all suffered from BIBD; it is therefore not possible to comment on its potential pathogenicity and contribution to the observed changes in the present case material.


Asunto(s)
Arenaviridae , Boidae/virología , Proteínas Virales , Animales , Arenaviridae/clasificación , Arenaviridae/genética , Arenaviridae/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo
10.
Viruses ; 11(8)2019 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-31416162

RESUMEN

A metatranscriptomic study of RNA viruses in cold-blooded vertebrates identified two related viruses from frogfish (Antennarius striatus) that represent a new genus Antennavirus in the family Arenaviridae (Order: Bunyavirales). Computational analyses were used to identify features common to class I viral fusion proteins (VFPs) in antennavirus glycoproteins, including an N-terminal fusion peptide, two extended alpha-helices, an intrahelical loop, and a carboxyl terminal transmembrane domain. Like mammarenavirus and hartmanivirus glycoproteins, the antennavirus glycoproteins have an intracellular zinc-binding domain and a long virion-associated stable signal peptide (SSP). The glycoproteins of reptarenaviruses are also class I VFPs, but do not contain zinc-binding domains nor do they encode SSPs. Divergent evolution from a common progenitor potentially explains similarities of antennavirus, mammarenavirus, and hartmanivirus glycoproteins, with an ancient recombination event resulting in a divergent reptarenavirus glycoprotein.


Asunto(s)
Infecciones por Arenaviridae/veterinaria , Arenaviridae/metabolismo , Enfermedades de los Peces/virología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales de Fusión/metabolismo , Zinc/metabolismo , Secuencia de Aminoácidos , Animales , Arenaviridae/química , Arenaviridae/genética , Infecciones por Arenaviridae/virología , Filogenia , Dominios Proteicos , Señales de Clasificación de Proteína , Proteómica , Alineación de Secuencia , Proteínas del Envoltorio Viral/genética , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/genética
11.
Virol Sin ; 31(5): 380-394, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27562602

RESUMEN

Mammarenaviruses, including lethal pathogens such as Lassa virus and Junín virus, can cause severe hemorrhagic fever in humans. Entry is a key step for virus infection, which starts with binding of the envelope glycoprotein (GP) to receptors on target cells and subsequent fusion of the virus with target cell membranes. The GP precursor is synthesized as a polypeptide, and maturation occurs by two cleavage events, yielding a tripartite GP complex (GPC) formed by a stable signal peptide (SSP), GP1 and GP2. The unique retained SSP interacts with GP2 and plays essential roles in virion maturation and infectivity. GP1 is responsible for binding to the cell receptor, and GP2 is a class I fusion protein. The native structure of the tripartite GPC is unknown. GPC is critical for the receptor binding, membrane fusion and neutralization antibody recognition. Elucidating the molecular mechanisms underlining the structure-function relationship of the three subunits is the key for understanding their function and can facilitate novel avenues for combating virus infections. This review summarizes the basic aspects and recent research of the structure-function relationship of the three subunits. We discuss the structural basis of the receptor-binding domain in GP1, the interaction between SSP and GP2 and its role in virion maturation and membrane fusion, as well as the mechanism by which glycosylation stabilizes the GPC structure and facilitates immune evasion. Understanding the molecular mechanisms involved in these aspects will contribute to the development of novel vaccines and treatment strategies against mammarenaviruses infection.


Asunto(s)
Infecciones por Arenaviridae/virología , Arenaviridae/metabolismo , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Animales , Arenaviridae/química , Arenaviridae/genética , Humanos , Proteínas del Envoltorio Viral/genética
13.
Curr Mol Med ; 5(8): 735-51, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16375709

RESUMEN

Arenaviruses merit significant attention both as tractable model systems to study acute and persistent viral infections, and as clinically important human pathogens. Evidence indicates that LCMV remains present in the USA and Europe and capable of causing significant morbidity in infected individuals, likely being a neglected human pathogen. Moreover, new arenaviruses are being discovered in the Americas on the average of one every three years, with some of them causing severe hemorrhagic fever. In addition, weaponized forms of these viruses pose a real threat as agents of bioterrorism. Therefore, it is important to develop effective vaccines and better antiviral drugs to combat the dual threats of naturally occurring and intentionally introduced Arenavirus infections. The development of arenavirus reverse genetic systems is allowing investigators to conduct a detailed molecular characterization of the viral cis-acting signals and trans-acting factors that control each of the steps of the Arenavirus life cycle, including RNA synthesis, packaging and budding. We will discuss how this new knowledge is facilitating the establishment of novel assays to identify and characterize compounds capable of interfering with specific steps of the virus life cycle. Likewise, the ability to generate predetermined specific mutations within the arenavirus genome, and analyze their phenotypic expression, would significantly contribute to the elucidation of arenavirus-host interactions, including the bases of their ability to persist, as well as to cause severe HF (hemorrhagic fever) disease in humans. These approaches could also lead to the development of novel potent and safe Arenavirus vaccines.


Asunto(s)
Anticuerpos Antivirales/uso terapéutico , Antivirales/farmacología , Infecciones por Arenaviridae/tratamiento farmacológico , Arenaviridae/efectos de los fármacos , Virus de la Coriomeningitis Linfocítica/efectos de los fármacos , Vacunas Virales , Arenaviridae/genética , Arenaviridae/metabolismo , Infecciones por Arenaviridae/epidemiología , Infecciones por Arenaviridae/prevención & control , Humanos , Virus de la Coriomeningitis Linfocítica/genética , Virus de la Coriomeningitis Linfocítica/metabolismo
14.
J Virol ; 79(22): 14282-96, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16254363

RESUMEN

The receptor for lymphocytic choriomeningitis virus (LCMV), the human pathogenic Lassa fever virus (LFV), and clade C New World arenaviruses is alpha-dystroglycan (alpha-DG), a cell surface receptor for proteins of the extracellular matrix (ECM). Specific posttranslational modification of alpha-DG by the glycosyltransferase LARGE is critical for its function as an ECM receptor. In the present study, we show that LARGE-dependent modification is also crucial for alpha-DG's function as a cellular receptor for arenaviruses. Virus binding involves the mucin-type domain of alpha-DG and depends on modification by LARGE. A crucial role of the LARGE-dependent glycosylation of alpha-DG for virus binding is found for several isolates of LCMV, LFV, and the arenaviruses Mobala and Oliveros. Since the posttranslational modification by LARGE is crucial for alpha-DG recognition by both arenaviruses and the host-derived ligand laminin, it also influences competition between virus and laminin for alpha-DG. Hence, LARGE-dependent glycosylation of alpha-DG has important implications for the virus-host cell interaction and the pathogenesis of LFV in humans.


Asunto(s)
Arenaviridae/fisiología , ARN Polimerasas Dirigidas por ADN/metabolismo , Distroglicanos/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Arenaviridae/metabolismo , Secuencia de Bases , Cruzamientos Genéticos , Cartilla de ADN , Femenino , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Virales/metabolismo
15.
Ann Inst Pasteur Virol ; 139(3): 277-83, 1988.
Artículo en Inglés | MEDLINE | ID: mdl-2849965

RESUMEN

We studied the binding of Junin virus (Arenaviridae) glycoproteins, G1 and G2, to two insolubilized lectins. The results showed that mannose, N-acetyl-glucosamine and galactose residues were exposed on G2, while only the latter predominated on G1. Heterogeneity of carbohydrate chains was found in G2, the only glycoprotein that was iodinated by the lactoperoxidase method.


Asunto(s)
Arenaviridae/metabolismo , Arenavirus del Nuevo Mundo/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Animales , Cromatografía de Afinidad , Lactoperoxidasa/metabolismo , Lectinas/metabolismo , Células Vero
17.
Acta Virol ; 29(2): 97-103, 1985 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-2860803

RESUMEN

Analysis by immune precipitation and sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) of intracellular [35S]-methionine-labelled specific Junín virus polypeptides demonstrated synthesis of the nucleoprotein (Np64) between 24 to 96 hr post-infection (p.i.). Two glycoproteins with apparent molecular weights (Mr) of 72,000 (Gp72) and 38,000 (Gp38) were detected by pulse labelling with [35S]-methionine or sugar precursor at about 48 hr p.i. The rate of synthesis of Gp38 increased up to 96 hr p.i. with concomitant decrease of Gp72. During pulse-chase experiments, Gp72 was cleaved to Gp38. These results suggest that Gp72 may be a precursor of the envelope structural glycoprotein. Using sera to virion fractions we found that Gp72 and Gp38 did not share antigenic determinants with Np64. We also identified a 200,000 Mr polypeptide (P200) in infected cells. The possibility that P200, Gp72 and Np64 are virus coded products is discussed.


Asunto(s)
Arenaviridae/metabolismo , Arenavirus del Nuevo Mundo/metabolismo , Biosíntesis de Péptidos , Proteínas Virales/biosíntesis , Animales , Arenavirus del Nuevo Mundo/genética , Línea Celular , Cricetinae , Glicoproteínas/biosíntesis , ARN Viral/análisis , Proteínas Virales/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...