Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Immunother Cancer ; 12(4)2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38631709

RESUMEN

BACKGROUND: Engineered arenavirus vectors have recently been developed to leverage the body's immune system in the fight against chronic viral infections and cancer. Vectors based on Pichinde virus (artPICV) and lymphocytic choriomeningitis virus (artLCMV) encoding a non-oncogenic fusion protein of human papillomavirus (HPV)16 E6 and E7 are currently being tested in patients with HPV16+ cancer, showing a favorable safety and tolerability profile and unprecedented expansion of tumor-specific CD8+ T cells. Although the strong antigen-specific immune response elicited by artLCMV vectors has been demonstrated in several preclinical models, PICV-based vectors are much less characterized. METHODS: To advance our understanding of the immunobiology of these two vectors, we analyzed and compared their individual properties in preclinical in vivo and in vitro systems. Immunogenicity and antitumor effect of intratumoral or intravenous administration of both vectors, as well as combination with NKG2A blockade, were evaluated in naïve or TC-1 mouse tumor models. Flow cytometry, Nanostring, and histology analysis were performed to characterize the tumor microenvironment (TME) and T-cell infiltrate following treatment. RESULTS: Despite being phylogenetically distant, both vectors shared many properties, including preferential infection and activation of professional antigen-presenting cells, and induction of potent tumor-specific CD8+ T-cell responses. Systemic as well as localized treatment induced a proinflammatory shift in the TME, promoting the infiltration of inducible T cell costimulator (ICOS)+CD8+ T cells capable of mediating tumor regression and prolonging survival in a TC-1 mouse tumor model. Still, there was evidence of immunosuppression built-up over time, and increased expression of H2-T23 (ligand for NKG2A T cell inhibitory receptor) following treatment was identified as a potential contributing factor. NKG2A blockade improved the antitumor efficacy of artARENA vectors, suggesting a promising new combination approach. This demonstrates how detailed characterization of arenavirus vector-induced immune responses and TME modulation can inform novel combination therapies. CONCLUSIONS: The artARENA platform represents a strong therapeutic vaccine approach for the treatment of cancer. The induced antitumor immune response builds the backbone for novel combination therapies, which warrant further investigation.


Asunto(s)
Arenavirus , Neoplasias , Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Humanos , Ratones , Animales , Linfocitos T CD8-positivos , Proteínas E7 de Papillomavirus , Arenavirus/metabolismo , Neoplasias/terapia , Modelos Animales de Enfermedad , Terapia de Inmunosupresión , Microambiente Tumoral
2.
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
3.
Viruses ; 13(6)2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34070281

RESUMEN

Arenaviruses cause chronic and asymptomatic infections in their natural host, rodents, and several arenaviruses cause severe hemorrhagic fever that has a high mortality in infected humans, seriously threatening public health. There are currently no FDA-licensed drugs available against arenaviruses; therefore, it is important to develop novel antiviral strategies to combat them, which would be facilitated by a detailed understanding of the interactions between the viruses and their hosts. To this end, we performed a transcriptomic analysis on cells infected with arenavirus lymphocytic choriomeningitis virus (LCMV), a neglected human pathogen with clinical significance, and found that the signal transducer and activator of transcription 3 (STAT3) signaling pathway was activated. A further investigation indicated that STAT3 could be activated by the RNA-dependent RNA polymerase L protein (Lp) of LCMV. Our functional analysis found that STAT3 cannot affect LCMV multiplication in A549 cells. We also found that STAT3 was activated by the Lp of Mopeia virus and Junin virus, suggesting that this activation may be conserved across certain arenaviruses. Our study explored the interactions between arenaviruses and STAT3, which may help us to better understand the molecular and cell biology of arenaviruses.


Asunto(s)
Arenavirus/enzimología , Arenavirus/metabolismo , Interacciones Huésped-Patógeno , ARN Polimerasa Dependiente del ARN/genética , Factor de Transcripción STAT3/genética , Transducción de Señal/genética , Células A549 , Arenavirus/genética , Arenavirus/patogenicidad , Línea Celular , Células HEK293 , Células HeLa , Humanos , ARN Polimerasa Dependiente del ARN/metabolismo , Transducción de Señal/fisiología , Replicación Viral
4.
Bioorg Med Chem Lett ; 41: 127983, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33965007

RESUMEN

We identified and explored the structure-activity relationship (SAR) of a novel heterocyclic chemical series of arenavirus cell entry inhibitors. Optimized lead compounds, including diphenyl-substituted imidazo[1,2-a]pyridines, benzimidazoles, and benzotriazoles exhibited low to sub-nanomolar potency against both pseudotyped and infectious Old and New World arenaviruses, attractive metabolic stability in human and most nonhuman liver microsomes as well as a lack of hERG K + channel or CYP enzyme inhibition. Moreover, the straightforward synthesis of several lead compounds (e.g., the simple high yield 3-step synthesis of imidazo[1,2-a]pyridine 37) could provide a cost-effective broad-spectrum arenavirus therapeutic that may help to minimize the cost-prohibitive burdens associated with treatments for emerging viruses in economically challenged geographical settings.


Asunto(s)
Antivirales/farmacología , Arenavirus/efectos de los fármacos , Descubrimiento de Drogas , Compuestos Heterocíclicos/farmacología , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Antivirales/síntesis química , Antivirales/química , Arenavirus/metabolismo , Relación Dosis-Respuesta a Droga , Compuestos Heterocíclicos/síntesis química , Compuestos Heterocíclicos/química , Humanos , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Relación Estructura-Actividad , Proteínas del Envoltorio Viral/metabolismo
5.
FEBS Open Bio ; 11(4): 1076-1083, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33534950

RESUMEN

Arenaviruses are enveloped viruses containing a segmented, negative, and ambisense single-stranded RNA genome wrapped with a nucleoprotein (NP). The NP is the most abundant viral protein in infected cells and plays a critical role in both replication/transcription and virion assembly. The NP associates with RNA to form a ribonucleoprotein (RNP) complex, and this implies self-assembly while the exact structure of this polymer is not yet known. Here, we report a measurement of the full-length Mopeia virus NP by negative stain transmission electron microscopy. We observed RNP complex particles with diameter 15 ± 1 nm as well as symmetric circular heptamers of the same diameter, consistent with previous observations.


Asunto(s)
Arenavirus , Nucleoproteínas/química , Multimerización de Proteína , Proteínas Virales/química , Secuencia de Aminoácidos , Arenavirus/metabolismo , Modelos Moleculares , Nucleoproteínas/metabolismo , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas de Unión al ARN/química , Proteínas Recombinantes , Proteínas Virales/metabolismo , Proteínas Virales/ultraestructura
6.
Bioorg Med Chem Lett ; 29(22): 126620, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31537423

RESUMEN

Old World (Africa) and New World (South America) arenaviruses are associated with human hemorrhagic fevers. Efforts to develop small molecule therapeutics have yielded several chemical series including the 4-acyl-1,6-dialkylpiperazin-2-ones. Herein, we describe an extensive exploration of this chemotype. In initial Phase I studies, R1 and R4 scanning libraries were assayed to identify potent substituents against Old World (Lassa) virus. In subsequent Phase II studies, R6 substituents and iterative R1, R4 and R6 substituent combinations were evaluated to obtain compounds with improved Lassa and New World (Machupo, Junin, and Tacaribe) arenavirus inhibitory activity, in vitro human liver microsome metabolic stability and aqueous solubility.


Asunto(s)
Antivirales/farmacología , Arenavirus/efectos de los fármacos , Piperazinas/farmacología , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Antivirales/síntesis química , Antivirales/química , Arenavirus/metabolismo , Relación Dosis-Respuesta a Droga , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Piperazinas/síntesis química , Piperazinas/química , Relación Estructura-Actividad , Proteínas del Envoltorio Viral/metabolismo
7.
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
8.
Curr Opin Virol ; 31: 52-58, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29843991

RESUMEN

The structure of a prefusion arenavirus GPC was enigmatic for many years, owing to the metastable and non-covalent nature of the association between the receptor binding and fusion subunits. Recent engineering efforts to stabilize the glycoprotein of the Old World arenavirus Lassa in a native, yet cleaved state, allowed the first structure of any arenavirus prefusion GPC trimer to be determined. Comparison of this structure with the structures of other arenavirus glycoprotein subunits reveals surprising findings: that the receptor binding subunit, GP1, of Lassa virus is conformationally labile, while the GP1 subunit of New World arenaviruses is not, and that the arenavirus GPC adopts a trimeric state unlike other glycoproteins with similar fusion machinery. Structural analysis, combined with recent biochemical data regarding antibody epitopes and receptor binding requirements, provides a basis for rational vaccine design.


Asunto(s)
Epítopos/inmunología , Glicoproteínas/química , Glicoproteínas/metabolismo , Virus Lassa/química , Arenavirus/metabolismo , Humanos , Virus Lassa/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus
9.
Methods Mol Biol ; 1604: 157-167, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28986831

RESUMEN

For many viruses that enter their target cells through pH-dependent fusion of the viral and endosomal membranes, cell-cell fusion assays can provide an experimental platform for investigating the structure-function relationships that promote envelope glycoprotein membrane-fusion activity. Typically, these assays employ effector cells expressing the recombinant envelope glycoprotein on the cell surface and target cells engineered to quantitatively report fusion with the effector cell. In the protocol described here, Vero cells are transfected with a plasmid encoding the arenavirus envelope glycoprotein complex GPC and infected with the vTF7-3 vaccinia virus expressing the bacteriophage T7 RNA polymerase. These effector cells are mixed with target cells infected with the vCB21R-lacZ vaccinia virus encoding a ß-galactosidase reporter under the control of the T7 promoter. Cell-cell fusion is induced upon exposure to low-pH medium (pH 5.0), and the resultant expression of the ß-galactosidase reporter is quantitated using a chemiluminescent substrate. We have utilized this robust microplate cell-cell fusion assay extensively to study arenavirus entry and its inhibition by small-molecule fusion inhibitors.


Asunto(s)
Arenavirus/metabolismo , Bioensayo/métodos , Endosomas/metabolismo , Glicoproteínas/metabolismo , Animales , Chlorocebus aethiops , Concentración de Iones de Hidrógeno , Fusión de Membrana/fisiología , Células Vero , Proteínas del Envoltorio Viral/metabolismo , beta-Galactosidasa/metabolismo
10.
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
11.
Methods Mol Biol ; 1604: 179-188, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28986833

RESUMEN

We describe methods to express the nucleoprotein (NP) of Lassa fever virus (LASV) in E. coli, to purify and crystallize it using the sitting-drop vapor diffusion method. The crystals were screened using Rigaku micro-007 X-ray generator and a dataset was collected at a resolution of 2.36 Å. The crystals belong to space group P3, with the unit cell parameters a = b = 176.35 Å, c = 56.40 Å, α = ß = 90°, and γ = 120°. Using the X-ray diffraction method, we constructed a three-dimensional structure of the LASV NP that should aid in the development of novel therapeutic strategies against this virus, for which vaccine and effective treatment modalities are currently unavailable.


Asunto(s)
Fiebre de Lassa/metabolismo , Fiebre de Lassa/virología , Virus Lassa/metabolismo , Nucleoproteínas/química , Nucleoproteínas/metabolismo , Arenavirus/genética , Arenavirus/metabolismo , Escherichia coli/metabolismo , Virus Lassa/genética , Nucleoproteínas/genética , Proteínas Virales/metabolismo , Difracción de Rayos X
12.
Methods Mol Biol ; 1604: 189-200, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28986834

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 (GPC). The NP encapsidates viral genome, is required for viral transcription and replication, and acts as a type I interferon (IFN) antagonist. This article describes assays to demonstrate that NP contains 3'-5' exoribonuclease (RNase) activity to degrade modeled RNA of the pathogen-associated molecular pattern type and suppresses the IFNß promoter-driven luciferase reporter gene. The minigenomic (MG) assay is used to assess the role of NP in replicating and transcribing a viral promoter-driven luciferase reporter gene. These powerful assays demonstrate the versatility of NP in mediating viral replication as well as in modulating host innate immune responses.


Asunto(s)
Arenavirus/metabolismo , Interferón Tipo I/metabolismo , Nucleoproteínas/metabolismo , Proteínas Virales/metabolismo , Animales , Arenavirus/genética , Genoma Viral/genética , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Inmunidad Innata/genética , Inmunidad Innata/fisiología , Nucleoproteínas/genética , ARN Viral/genética , Proteínas Virales/genética
13.
Methods Mol Biol ; 1604: 209-215, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28986836

RESUMEN

Independent expression of the VP40 or Z matrix proteins of filoviruses (marburgviruses and ebolaviruses) and arenaviruses (Lassa fever and Junín), respectively, gives rise to the production and release of virus-like particles (VLPs) that are morphologically identical to infectious virions. We can detect and quantify VLP production and egress in mammalian cells by transient transfection, SDS-PAGE, Western blotting, and live cell imaging techniques such as total internal reflection fluorescence (TIRF) microscopy. Since the VLP budding assay accurately mimics budding of infectious virus, this BSL-2 assay is safe and useful for the interrogation of both viral and host determinants required for budding and can be used as an initial screen to identify and validate small molecule inhibitors of virus release and spread.


Asunto(s)
Fiebre Hemorrágica Ebola/metabolismo , Liberación del Virus/fisiología , Animales , Arenavirus/genética , Arenavirus/metabolismo , Western Blotting , Ebolavirus/genética , Ebolavirus/metabolismo , Electroforesis en Gel de Poliacrilamida , Filoviridae/genética , Filoviridae/metabolismo , Fiebre Hemorrágica Ebola/genética , Humanos , Virus Junin/genética , Virus Junin/metabolismo , Fiebre de Lassa/virología , Marburgvirus/genética , Marburgvirus/metabolismo , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo , Liberación del Virus/genética
14.
Methods Mol Biol ; 1604: 217-227, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28986837

RESUMEN

The smallest arenaviral protein is the zinc-finger protein (Z) that belongs to the RING finger protein family. Z serves as a main component required for virus budding from the membrane of the infected cells through self-oligomerization, a process that can be aided by the viral nucleoprotein (NP) to form the viral matrix of progeny virus particles. Z has also been shown to be essential for mediating viral transcriptional repression activity by locking the L polymerase onto the viral promoter in a catalytically inactive state, thus limiting viral replication. The Z protein has also recently been shown to inhibit the type I interferon-induction pathway by directly binding to the intracellular pathogen-sensor proteins RIG-I and MDA5, and thus inhibiting their normal functions. This chapter describes several assays used to examine the important roles of the arenaviral Z protein in mediating virus budding (i.e., either Z self-budding or NP-Z budding activities), viral transcriptional inhibition in a viral minigenome (MG) assay, and type I IFN suppression in an IFN-ß promoter-mediated luciferase reporter assay.


Asunto(s)
Arenavirus/metabolismo , Liberación del Virus/fisiología , Replicación Viral/fisiología , Arenavirus/genética , Interferón beta/metabolismo , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , Virus Pichinde/genética , Virus Pichinde/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Liberación del Virus/genética , Replicación Viral/genética
15.
Acta Crystallogr D Struct Biol ; 73(Pt 8): 641-649, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28777079

RESUMEN

The Arenaviridae family is one of the two RNA viral families that encode a 3'-5' exonuclease in their genome. An exonuclease domain is found in the Arenaviridae nucleoprotein and targets dsRNA specifically. This domain is directly involved in suppression of innate immunity in the host cell. Like most phosphate-processing enzymes, it requires a divalent metal ion such as Mg2+ (or Mn2+) as a cofactor to catalyse nucleotide-cleavage and nucleotide-transfer reactions. On the other hand, calcium (Ca2+) inhibits this enzymatic activity, in spite of the fact that Mg2+ and Ca2+ present comparable binding affinities and biological availabilities. Here, the molecular and structural effects of the replacement of magnesium by calcium and its inhibition mechanism for phosphodiester cleavage, an essential reaction in the viral process of innate immunity suppression, are studied. Biochemical data and high-resolution structures of the Mopeia virus exonuclease domain complexed with each ion are reported for the first time. The consequences of the ion swap for the stability of the protein, the catalytic site and the functional role of a specific metal ion in enabling the catalytic cleavage of a dsRNA substrate are outlined.


Asunto(s)
Arenavirus/química , Arenavirus/enzimología , Exonucleasas/química , Proteínas de la Nucleocápside/química , Nucleoproteínas/química , Infecciones por Arenaviridae/virología , Arenavirus/metabolismo , Sitios de Unión , Calcio/metabolismo , Dominio Catalítico , Cationes Bivalentes/metabolismo , Cristalización , Cristalografía por Rayos X , Exonucleasas/metabolismo , Magnesio/metabolismo , Manganeso/metabolismo , Simulación del Acoplamiento Molecular , Proteínas de la Nucleocápside/metabolismo , Nucleoproteínas/metabolismo , Dominios Proteicos , ARN Viral/metabolismo
16.
Glycobiology ; 27(9): 806-819, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28810660

RESUMEN

The post-translational glycosylation of select proteins by O-linked mannose (O-mannose or O-man) is a conserved modification from yeast to humans and has been shown to be necessary for proper development and growth. The most well studied O-mannosylated mammalian protein is α-dystroglycan (α-DG). Hypoglycosylation of α-DG results in varying severities of congenital muscular dystrophies, cancer progression and metastasis, and inhibited entry and infection of certain arenaviruses. Defects in the gene products responsible for post-translational modification of α-DG, primarily glycosyltransferases, are the basis for these diseases. The multitude of clinical phenotypes resulting from defective O-mannosylation highlights the biomedical significance of this unique modification. Elucidation of the various O-mannose biosynthetic pathways is imperative to understanding a broad range of human diseases and for the development of novel therapeutics. In this review, we will focus on recent discoveries delineating the various enzymes, structures and functions associated with O-mannose-initiated glycoproteins. Additionally, we discuss current gaps in our knowledge of mammalian O-mannosylation, discuss the evolution of this pathway, and illustrate the utility and limitations of model systems to study functions of O-mannosylation.


Asunto(s)
Distroglicanos/química , Glicosiltransferasas/metabolismo , Manosa/metabolismo , Distrofias Musculares/metabolismo , Neoplasias/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Arenavirus/metabolismo , Distroglicanos/genética , Distroglicanos/metabolismo , Evolución Molecular , Glicosilación , Glicosiltransferasas/genética , Humanos , Mamíferos , Manosa/química , Distrofias Musculares/genética , Distrofias Musculares/patología , Neoplasias/genética , Neoplasias/patología , Receptores Virales/química , Receptores Virales/genética , Receptores Virales/metabolismo
17.
PLoS Pathog ; 13(1): e1006073, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28076397

RESUMEN

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.


Asunto(s)
Arenavirus/clasificación , Arenavirus/metabolismo , Reservorios de Enfermedades/virología , Murinae/virología , Enfermedades de los Roedores/virología , Animales , Arenavirus/fisiología , Humanos , Fiebre de Lassa/virología , Virus Lassa/fisiología , Filogeografía , Especificidad de la Especie , Tanzanía
18.
J Virol ; 90(22): 10259-10270, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27605671

RESUMEN

The family Arenaviridae includes several important human pathogens that can cause severe hemorrhagic fever and greatly threaten public health. As a major component of the innate immune system, the RLR/MAVS signaling pathway is involved in recognizing viral components and initiating antiviral activity. It has been reported that arenavirus infection can suppress the innate immune response, and NP and Z proteins of pathogenic arenaviruses can disrupt RLR/MAVS signaling, thus inhibiting production of type I interferon (IFN-I). However, recent studies have shown elevated IFN-I levels in certain arenavirus-infected cells. The mechanism by which arenavirus infection induces IFN-I responses remains unclear. In this study, we determined that the L polymerase (Lp) of Mopeia virus (MOPV), an Old World (OW) arenavirus, can activate the RLR/MAVS pathway and thus induce the production of IFN-I. This activation is associated with the RNA-dependent RNA polymerase activity of Lp. This study provides a foundation for further studies of interactions between arenaviruses and the innate immune system and for the elucidation of arenavirus pathogenesis. IMPORTANCE: Distinct innate immune responses are observed when hosts are infected with different arenaviruses. It has been widely accepted that NP and certain Z proteins of arenaviruses inhibit the RLR/MAVS signaling pathway. The viral components responsible for the activation of the RLR/MAVS signaling pathway remain to be determined. In the current study, we demonstrate for the first time that the Lp of MOPV, an OW arenavirus, can activate the RLR/MAVS signaling pathway and thus induce the production of IFN-I. Based on our results, we proposed that dynamic interactions exist among Lp-produced RNA, NP, and the RLR/MAVS signaling pathway, and the outcome of these interactions may determine the final IFN-I response pattern: elevated or reduced. Our study provides a possible explanation for how IFN-I can become activated during arenavirus infection and may help us gain insights into the interactions that form between different arenavirus components and the innate immune system.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Infecciones por Arenaviridae/metabolismo , Arenavirus del Viejo Mundo/metabolismo , Transducción de Señal/fisiología , Proteínas Virales/metabolismo , Animales , Infecciones por Arenaviridae/inmunología , Infecciones por Arenaviridae/virología , Arenavirus/inmunología , Arenavirus/metabolismo , Arenavirus del Viejo Mundo/inmunología , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , ARN Polimerasas Dirigidas por ADN/metabolismo , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno/inmunología , Interacciones Huésped-Patógeno/fisiología , Humanos , Inmunidad Innata/inmunología , Interferón Tipo I/metabolismo , Células Vero
19.
J Virol ; 89(10): 5734-8, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25762740

RESUMEN

In this study, we document that efficient interaction between arenavirus nucleoprotein (NP) and RNA-dependent RNA polymerase (L protein), the two trans-acting viral factors required for both virus RNA replication and gene transcription, requires the presence of virus-specific RNA sequences located within the untranslated 5' and 3' termini of the viral genome.


Asunto(s)
Arenavirus/metabolismo , Nucleocápside/metabolismo , Nucleoproteínas/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas Virales/metabolismo , Arenavirus/genética , Genoma Viral , Células HEK293 , Humanos , Virus de la Coriomeningitis Linfocítica/genética , Virus de la Coriomeningitis Linfocítica/fisiología , Nucleocápside/genética , Nucleoproteínas/genética , ARN no Traducido/genética , ARN no Traducido/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , ARN Polimerasa Dependiente del ARN/genética , Proteínas Virales/genética , Replicación Viral
20.
J Mol Biol ; 426(7): 1452-68, 2014 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-24333483

RESUMEN

Fusion of the viral and host cell membranes is a necessary first step for infection by enveloped viruses and is mediated by the envelope glycoprotein. The transmembrane subunits from the structurally defined "class I" glycoproteins adopt an α-helical "trimer-of-hairpins" conformation during the fusion pathway. Here, we present our studies on the envelope glycoprotein transmembrane subunit, GP2, of the CAS virus (CASV). CASV was recently identified from annulated tree boas (Corallus annulatus) with inclusion body disease and is implicated in the disease etiology. We have generated and characterized two protein constructs consisting of the predicted CASV GP2 core domain. The crystal structure of the CASV GP2 post-fusion conformation indicates a trimeric α-helical bundle that is highly similar to those of Ebola virus and Marburg virus GP2 despite CASV genome homology to arenaviruses. Denaturation studies demonstrate that the stability of CASV GP2 is pH dependent with higher stability at lower pH; we propose that this behavior is due to a network of interactions among acidic residues that would destabilize the α-helical bundle under conditions where the side chains are deprotonated. The pH-dependent stability of the post-fusion structure has been observed in Ebola virus and Marburg virus GP2, as well as other viruses that enter via the endosome. Infection experiments with CASV and the related Golden Gate virus support a mechanism of entry that requires endosomal acidification. Our results suggest that, despite being primarily arenavirus like, the transmembrane subunit of CASV is extremely similar to the filoviruses.


Asunto(s)
Arenavirus/metabolismo , Laminina/química , Secuencia de Aminoácidos , Ebolavirus/metabolismo , Datos de Secuencia Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...