Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Molecules ; 29(10)2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38792166

RESUMEN

Food-borne transmission is a recognized route for many viruses associated with gastrointestinal, hepatic, or neurological diseases. Therefore, it is essential to identify new bioactive compounds with broad-spectrum antiviral activity to exploit innovative solutions against these hazards. Recently, antimicrobial peptides (AMPs) have been recognized as promising antiviral agents. Indeed, while the antibacterial and antifungal effects of these molecules have been widely reported, their use as potential antiviral agents has not yet been fully investigated. Herein, the antiviral activity of previously identified or newly designed AMPs was evaluated against the non-enveloped RNA viruses, hepatitis A virus (HAV) and murine norovirus (MNV), a surrogate for human norovirus. Moreover, specific assays were performed to recognize at which stage of the viral infection cycle the peptides could function. The results showed that almost all peptides displayed virucidal effects, with about 90% of infectivity reduction in HAV or MNV. However, the decapeptide RiLK1 demonstrated, together with its antibacterial and antifungal properties, a notable reduction in viral infection for both HAV and MNV, possibly through direct interaction with viral particles causing their damage or hindering the recognition of cellular receptors. Hence, RiLK1 could represent a versatile antimicrobial agent effective against various foodborne pathogens including viruses, bacteria, and fungi.


Asunto(s)
Antivirales , Enfermedades Transmitidas por los Alimentos , Norovirus , Antivirales/farmacología , Antivirales/química , Animales , Enfermedades Transmitidas por los Alimentos/prevención & control , Enfermedades Transmitidas por los Alimentos/tratamiento farmacológico , Enfermedades Transmitidas por los Alimentos/virología , Norovirus/efectos de los fármacos , Humanos , Ratones , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/química , Virus de la Hepatitis A/efectos de los fármacos , Virosis/tratamiento farmacológico , Pruebas de Sensibilidad Microbiana
2.
bioRxiv ; 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38260524

RESUMEN

Non-enveloped viruses employ unique entry mechanisms to breach and infect host cells. Understanding these mechanisms is crucial for developing antiviral strategies. Prevailing perspective suggests that non-enveloped viruses release membrane lytic peptides to breach host membranes. However, the precise involvement of the viral capsid in this entry remains elusive. Our study presents direct observations elucidating the dynamically distinctive steps through which metastable reovirus capsids disrupt host lipid membranes as they uncoat into partially hydrophobic intermediate particles. Using both live cells and model membrane systems, our key finding is that reovirus capsids actively deform and permeabilize lipid membranes in a cholesterol-dependent process. Unlike membrane lytic peptides, these metastable viral capsids induce more extensive membrane perturbations, including budding, bridging between adjacent membranes, and complete rupture. Notably, cholesterol enhances subviral particle adsorption, resulting in the formation of pores equivalent to the capsid size. This cholesterol dependence is attributed to the lipid condensing effect, particularly prominent at intermediate cholesterol level. Furthermore, our results reveal a positive correlation between membrane disruption extent and efficiency of viral variants in establishing infection. This study unveils the crucial role of capsid-lipid interaction in non-enveloped virus entry, providing new insights into how cholesterol homeostasis influences virus infection dynamics.

3.
Microbiol Spectr ; : e0190823, 2023 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-37728342

RESUMEN

Ebola virus (EBOV) causes a hemorrhagic fever with fatality rates up to 90%. The EBOV entry process is complex and incompletely understood. Following attachment to host cells, EBOV is trafficked to late endosomes/lysosomes where its glycoprotein (GP) is processed to a 19-kDa form, which binds to the EBOV intracellular receptor Niemann-Pick type C1. We previously showed that the cathepsin protease inhibitor, E-64d, blocks infection by pseudovirus particles bearing 19-kDa GP, suggesting that further cathepsin action is needed to trigger fusion. This, however, has not been demonstrated directly. Since 19-kDa Ebola GP fusion occurs in late endosomes, we devised a system in which enriched late endosomes are used to prepare supported planar endosomal membranes (SPEMs), and fusion of fluorescent (pseudo)virus particles is monitored by total internal reflection fluorescence microscopy. We validated the system by demonstrating the pH dependencies of influenza virus hemagglutinin (HA)-mediated and Lassa virus (LASV) GP-mediated fusion. Using SPEMs, we showed that fusion mediated by 19-kDa Ebola GP is dependent on low pH, enhanced by Ca2+, and augmented by the addition of cathepsins. Subsequently, we found that E-64d inhibits full fusion, but not lipid mixing, mediated by 19-kDa GP, which we corroborated with the reversible cathepsin inhibitor VBY-825. Hence, we provide both gain- and loss-of-function evidence that further cathepsin action enhances the fusion activity of 19-kDa Ebola GP. In addition to providing new insights into how Ebola GP mediates fusion, the approach we developed employing SPEMs can now be broadly used for studies of virus and toxin entry through endosomes. IMPORTANCE Ebola virus is the causative agent of Ebola virus disease, which is severe and frequently lethal. EBOV gains entry into cells via late endosomes/lysosomes. The events immediately preceding fusion of the viral and endosomal membranes are incompletely understood. In this study, we report a novel in vitro system for studying virus fusion with endosomal membranes. We validated the system by demonstrating the low pH dependencies of influenza and Lassa virus fusion. Moreover, we show that further cathepsin B action enhances the fusion activity of the primed Ebola virus glycoprotein. Finally, this model endosomal membrane system should be useful in studying the mechanisms of bilayer breaching by other enveloped viruses, by non-enveloped viruses, and by acid-activated bacterial toxins.

4.
J Virol ; 97(8): e0075623, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37578227

RESUMEN

During entry, non-enveloped viruses penetrate a host membrane to cause infection, although how this is accomplished remains enigmatic. Polyomaviruses (PyVs) are non-enveloped DNA viruses that penetrate the endoplasmic reticulum (ER) membrane to reach the cytosol en route to the nucleus for infection. To penetrate the ER membrane, the prototype PyV simian virus 40 (SV40) induces formation of ER-escape sites, called foci, composed of repeating units of multi-tubular ER junctions where the virus is thought to exit. How SV40 triggers formation of the ER-foci harboring these multi-tubular ER junctions is unclear. Here, we show that the ER morphogenic atlastin 2 (ATL2) and ATL3 membrane proteins play critical roles in SV40 infection. Mechanistically, ATL3 mobilizes to the ER-foci where it deploys its GTPase-dependent membrane fusion activity to promote formation of multi-tubular ER junctions within the ER-foci. ATL3 also engages an SV40-containing membrane penetration complex. By contrast, ATL2 does not reorganize to the ER-foci. Instead, it supports the reticular ER morphology critical for the integrity of the ATL3-dependent membrane complex. Our findings illuminate how two host factors play distinct roles in the formation of an essential membrane penetration site for a non-enveloped virus. IMPORTANCE Membrane penetration by non-enveloped viruses, a critical infection step, remains enigmatic. The non-enveloped PyV simian virus 40 (SV40) penetrates the endoplasmic reticulum (ER) membrane to reach the cytosol en route for infection. During ER-to-cytosol membrane penetration, SV40 triggers formation of ER-associated structures (called ER-foci) that function as the membrane penetration sites. Here, we discover a role of the ATL ER membrane proteins-known to shape the ER morphology-during SV40-induced ER-foci formation. These findings illuminate how a non-enveloped virus hijacks host components to construct a membrane penetration structure.


Asunto(s)
Membranas Intracelulares , Chaperonas Moleculares , Membranas Intracelulares/metabolismo , Chaperonas Moleculares/metabolismo , Internalización del Virus , Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/metabolismo
5.
Viruses ; 15(7)2023 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-37515296

RESUMEN

Vesicle-encapsulated nonenveloped viruses are a recently recognized alternate form of nonenveloped viruses that can avoid immune detection and potentially increase systemic transmission. Avian orthoreoviruses (ARVs) are the leading cause of various disease conditions among birds and poultry. However, whether ARVs use cellular vesicle trafficking routes for egress and cell-to-cell transmission is still poorly understood. We demonstrated that fusogenic ARV-infected quail cells generated small (~100 nm diameter) extracellular vesicles (EVs) that contained electron-dense material when observed by transmission electron microscope. Cryo-EM tomography indicated that these vesicles did not contain ARV virions or core particles, but the EV fractions of OptiPrep gradients did contain a small percent of the ARV virions released from cells. Western blotting of detergent-treated EVs revealed that soluble virus proteins and the fusogenic p10 FAST protein were contained within the EVs. Notably, virus particles mixed with the EVs were up to 50 times more infectious than virions alone. These results suggest that EVs and perhaps fusogenic FAST-EVs could contribute to ARV virulence.


Asunto(s)
Vesículas Extracelulares , Orthoreovirus Aviar , Vesículas Extracelulares/metabolismo , Proteínas Virales/metabolismo
6.
Virology ; 585: 232-239, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37406580

RESUMEN

Viruses can be involved in respiratory disorders in horses, with limited therapeutic options. Citrate-complexed silver nanoparticles (C-AgNP) have shown bactericidal properties after in vitro nebulization. The aim of the present study was to assess the virucidal activity of C-AgNP after in vitro instillation or nebulization on equine herpesvirus-1 (EHV-1) and murine norovirus (MNV), the latter used as surrogate for small non-enveloped viruses. Both viruses were instilled or nebulized with C-AgNP of increasing concentrations, and titres were determined via TCID50 method. We demonstrated efficient inactivation of enveloped EHV-1 following instillation and nebulization of C-AgNP (infectivity losses of ≥ three orders of magnitude). While tenacious MNV was inactivated via 2000 ppm C-AgNP instillation, nebulized C-AgNP did not lead to reduction in MNV titres. Nebulization of C-AgNP may represent a novel virucidal therapeutic approach in horses. Further investigations are needed to assess its safety and effective concentrations for in vivo use.


Asunto(s)
Herpesvirus Équido 1 , Nanopartículas del Metal , Norovirus , Animales , Caballos , Ratones , Ácido Cítrico , Plata/farmacología , Norovirus/fisiología
7.
Virology ; 585: 139-144, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37343460

RESUMEN

The surface hydrophobicity of native or engineered non-enveloped viruses and virus-like particles (VLPs) is a key parameter regulating their fate in living and artificial aqueous systems. Its modulation is mainly depending on the structure and environment of particles. Nevertheless, unexplained variations have been reported between structurally similar viruses and with pH. This indicates that some modulating factors of their hydrophobicity remain to be identified. Herein we investigate the potential involvement of RNA cargo in the MS2 phage used as non-enveloped RNA virus model, by examining the SDS-induced electrophoretic mobility shift (SEMS) determined for native MS2 virions and corresponding RNA-free VLPs at various pH. Interestingly, the SEMS of VLPs was larger and more variable from pH 5 to 9 compared to native virions. These observations are discussed in term of RNA-dependent changes in surface hydrophobicity, suggesting that RNA cargo may be a major modulator/regulator of this viral parameter.


Asunto(s)
Levivirus , ARN Viral , Levivirus/genética , Levivirus/química , ARN Viral/genética , Interacciones Hidrofóbicas e Hidrofílicas
8.
Peptides ; 166: 171024, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37172781

RESUMEN

Viral epidemics are occurring frequently, and the COVID-19 viral pandemic has resulted in at least 6.5 million deaths worldwide. Although antiviral therapeutics are available, these may not have sufficient effect. The emergence of resistant or novel viruses requires new therapies. Cationic antimicrobial peptides are agents of the innate immune system that may offer a promising solution to viral infections. These peptides are gaining attention as possible therapies for viral infections or for use as prophylactic agents to prevent viral spread. This narrative review examines antiviral peptides, their structural features, and mechanism of activity. A total of 156 cationic antiviral peptides were examined for information of their mechanism of action against both enveloped and non-enveloped viruses. Antiviral peptides can be isolated from various natural sources or can be generated synthetically. The latter tend to be more specific and effective and can be made to have a broad spectrum of activity with minimal side effects. Their unique properties of being positively charged and amphipathic enable their main mode of action which is to target and disrupt viral lipid envelopes, thereby inhibiting viral entry and replication. This review offers a comprehensive summary of the current understanding of antiviral peptides, which could potentially aid in the design and creation of novel antiviral medications.


Asunto(s)
COVID-19 , Virosis , Virus , Humanos , Antivirales/farmacología , Antivirales/uso terapéutico , Péptidos Catiónicos Antimicrobianos/farmacología , Péptidos Catiónicos Antimicrobianos/uso terapéutico , Virosis/tratamiento farmacológico
9.
Curr Protoc ; 3(4): e716, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37039704

RESUMEN

Mammalian reoviruses are pathogens that cause gastrointestinal and respiratory infections. In humans, the mammalian reoviruses usually cause mild or subclinical disease, and they are ubiquitous, with most people mounting immunity at a young age. Reoviruses are prototypic representations of the Reoviridae family, which contains many highly pathogenic viruses. This article describes techniques for culturing mouse fibroblast L929 cell lines, the preferred cell line in which most mammalian reovirus studies take place. In addition, mammalian reovirus propagation, quantification, purification, and storage are described. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Propagation of mammalian reoviruses in cell culture from virus stocks Alternate Protocol 1: Large-scale propagation (and purification) of mammalian reoviruses in cell culture from virus stocks Basic Protocol 2: Quantification of mammalian reoviruses by plaque assay with neutral red staining Alternate Protocol 2: Quantification of mammalian reoviruses by plaque assay with crystal violet staining Basic Protocol 3: Storage of mammalian reoviruses Support Protocol 1: Growth and maintenance of mouse L929 cells Support Protocol 2: Plating L929 cells.


Asunto(s)
Orthoreovirus de los Mamíferos , Orthoreovirus , Reoviridae , Humanos , Animales , Ratones , Línea Celular , Técnicas de Cultivo de Célula/métodos , Mamíferos
10.
J Microbiol Immunol Infect ; 55(6 Pt 2): 1151-1158, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35450828

RESUMEN

BACKGROUND: In Taiwan, there were only 799 confirmed COVID-19 cases in 2020. The unique backdrop amidst a pandemic and promotion of nonpharmaceutical interventions generated some distinct changes in the epidemiology of common respiratory pathogens. In this study, we aimed to investigate the dynamic changes in respiratory pathogens in children during 2020. METHODS: We performed a retrospective cohort study at a tertiary hospital in southern Taiwan during 2020. Patients aged 0-18 years who visited the pediatric emergency department were enrolled. Children who presented with clinical symptoms (fever or respiratory illness) and received nasopharyngeal swabs for multiplex polymerase chain reaction (PCR) were included in our analysis. We also compared respiratory syncytial virus (RSV) trends from previous years by PCR and lateral flow immunochromatographic assays from 2017 to 2020. RESULTS: A total of 120 children were tested. The overall detection rate was 55%. With strengthened restrictions, the detection rate dropped from 70% to 30%. However, non-enveloped viruses (rhinovirus/enterovirus and adenovirus) were in constant circulation. Upon easing prevention measures, the detection rate remained above 60%, and an outbreak of an enveloped virus (RSV and parainfluenza virus) was noted. Compared with 2017-2019, the cyclical RSV epidemic was delayed, with a large surge in late 2020. CONCLUSIONS: We observed a constant circulation of non-enveloped viruses when strict nonpharmaceutical interventions were employed and a delayed surge of enveloped viruses during the easing of restrictions. Continuous surveillance and monitoring of the evolutionary dynamics of respiratory viruses is important, while easing restrictions requires balanced judgment.


Asunto(s)
COVID-19 , Virus Sincitial Respiratorio Humano , Infecciones del Sistema Respiratorio , Virus , Niño , Humanos , Lactante , Estudios Retrospectivos , Incidencia , Taiwán/epidemiología , COVID-19/epidemiología , Reacción en Cadena de la Polimerasa Multiplex/métodos
11.
Environ Sci Pollut Res Int ; 29(19): 28565-28571, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34988790

RESUMEN

Phycoremediation of swine wastewater is a promising treatment since it efficiently removes nutrients and contaminants and, simultaneously, its biomass can be harvested and used to obtain a wide range of valuable compounds and metabolites. In this context, biomass microalgae were investigated for the phycoremediation of swine wastewater, and biomass extracts for its virucidal effect against enveloped and non-enveloped viruses. Microalgae were cultivated in a pilot scale bioreactor fed with swine wastewater as the growth substrate. Hexane, dichloromethane, and methanol were used to obtain the microalgae extracts. Extracts were tested for virucidal potential against HSV-1 and HAdV-5. Virucidal assays were conducted at temperatures that emulate environmental conditions (21 °C) and body temperature (37 °C). The maximum production of microalgae biomass reached a concentration of 318.5 ± 23.6 mgDW L-1. The results showed that phycoremediation removed 100% of ammonia-N and phosphate-P, with rates (k1) of 0.218 ± 0.013 and 0.501 ± 0.038 (day-1), respectively. All microalgae extract reduced 100% of the infectious capacity of HSV-1. The microalgae extracts with dichloromethane and methanol showed inhibition activities at the lowest concentration (3.125 µg mL-1). Virucidal assays against HAdV-5 using microalgae extract of hexane and methanol inhibited the infectious capacity of the virus by 70% at all concentrations tested at 37 °C. At a concentration of 12.5 µg mL-1, the dichloromethane microalgae extract reduced 50-80% of the infectious capacity of HAdV-5, also at 37 °C. Overall, the results suggest that the microalgae can be an attractive source of feedstock biomass for the exploration of alternative virucidal compounds.


Asunto(s)
Chlorella , Microalgas , Animales , Biomasa , Hexanos , Metanol/metabolismo , Cloruro de Metileno , Microalgas/metabolismo , Nitrógeno/análisis , Extractos Vegetales/metabolismo , Porcinos , Aguas Residuales
12.
Virology ; 568: 23-30, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35077959

RESUMEN

The hydrophobicity of virions is a major physicochemical parameter regulating their dissemination in humans and the environment. But knowledge about potential factors modulating virion hydrophobicity is limited due to the lack of suitable quantifying methods. It has been recently shown that sodium dodecyl-sulfate (SDS) labels capsid hydrophobic domains in capillary zone electrophoresis of non-enveloped virions, altering their electrophoretic mobility (µ) in proportion to their hydrophobicity. This was exploited here to quantify the hydrophobicity of GA, Qß and MS2 phages as a function of pH. By subtracting the native from the SDS-modified µ of phages, measured in the absence and presence of SDS, respectively, we defined a "hydrophobic index" increasing with virion hydrophobicity. Using this approach, we found that the virion hydrophobicity changes at a virion-specific pivotal pH. This procedure may be applied under various physicochemical conditions and to diverse non-enveloped virus families of significance to human health and the environment.


Asunto(s)
Electroforesis Capilar , Interacciones Hidrofóbicas e Hidrofílicas , Virión/química , Algoritmos , Secuencia de Aminoácidos , Bacteriófagos/química , Humanos , Modelos Teóricos , Dodecil Sulfato de Sodio , Proteínas Virales/química , Virión/aislamiento & purificación , Virión/ultraestructura
13.
Am J Infect Control ; 50(7): 764-771, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34864086

RESUMEN

BACKGROUND: The purpose of this study was to evaluate the virucidal activity of a new olanexidine-containing formulation for hand hygiene (olanexidine gluconate hand rub; OLG-HR) against non-enveloped viruses and to understand its mechanism of action. METHODS: The virucidal activities of OLG-HR against two strains of caliciviruses and three adenovirus serotypes were evaluated through suspension tests. Also, virus-like particles were used to predict the effect of olanexidine gluconate on virus particle structure. RESULTS: The results of suspension tests under conditions with and without interfering substances (1.5% BSA) indicated that OLG-HR had a broad-spectrum effect against non-enveloped viruses, and the virucidal effect was unaffected by organic contaminants. Furthermore, olanexidine inhibited the binding ability of virus-like particles to the binding receptor of human norovirus and increased the aggregation of virus-like particles in a dose-dependent manner. Transmission electron microscopy showed that the morphology of the virus-like particles was affected by exposure to olanexidine, indicating that the protein-denaturing effect of olanexidine gluconate caused the loss of receptor-binding capability of the viral capsid protein. CONCLUSIONS: This study suggests that olanexidine gluconate is a potential biological and environmental disinfectant against norovirus and adenovirus.


Asunto(s)
Antiinfecciosos Locales , Desinfectantes , Norovirus , Antiinfecciosos Locales/farmacología , Biguanidas , Desinfectantes/farmacología , Desinfección , Glucuronatos , Humanos
14.
Cell Rep ; 37(10): 110077, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34879280

RESUMEN

Viruses rearrange host membranes to support different entry steps. Polyomavirus simian virus 40 (SV40) reorganizes the endoplasmic reticulum (ER) membrane to generate focus structures that enable virus ER-to-cytosol escape, a decisive infection step. The molecular architecture of the ER exit site that might illuminate why it is ideally suited for membrane penetration is unknown. Here 3D focused ion beam scanning electron microscopy (FIB-SEM) reconstruction reveals that the ER focus structure consists of multi-tubular ER junctions where SV40 preferentially localizes, suggesting that tubular branch points are virus ER-to-cytosol penetration sites. Functional analysis demonstrates that lunapark-an ER membrane protein that typically stabilizes three-way ER junctions-relocates to the ER foci, where it supports focus formation, leading to SV40 ER escape and infection. Our results reveal how a virus repurposes the activity of an ER membrane protein to form a virus-induced ER substructure required for membrane escape and suggest that ER tubular junctions are vulnerable sites exploited by viruses for membrane penetration.


Asunto(s)
Citosol/virología , Retículo Endoplásmico/metabolismo , Membranas Intracelulares/metabolismo , Proteínas de la Membrana/metabolismo , Virus 40 de los Simios/metabolismo , Internalización del Virus , Animales , Línea Celular , Chlorocebus aethiops , Citosol/metabolismo , Retículo Endoplásmico/genética , Retículo Endoplásmico/ultraestructura , Retículo Endoplásmico/virología , Interacciones Huésped-Patógeno , Membranas Intracelulares/ultraestructura , Membranas Intracelulares/virología , Masculino , Proteínas de la Membrana/genética , Virus 40 de los Simios/patogenicidad , Virus 40 de los Simios/ultraestructura
15.
J Gen Virol ; 102(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33560198

RESUMEN

A long-standing paradigm in virology was that non-enveloped viruses induce cell lysis to release progeny virions. However, emerging evidence indicates that some non-enveloped viruses exit cells without inducing cell lysis, while others engage both lytic and non-lytic egress mechanisms. Enteric viruses are transmitted via the faecal-oral route and are important causes of a wide range of human infections, both gastrointestinal and extra-intestinal. Virus cellular egress, when fully understood, may be a relevant target for antiviral therapies, which could minimize the public health impact of these infections. In this review, we outline lytic and non-lytic cell egress mechanisms of non-enveloped enteric RNA viruses belonging to five families: Picornaviridae, Reoviridae, Caliciviridae, Astroviridae and Hepeviridae. We discuss factors that contribute to egress mechanisms and the relevance of these mechanisms to virion stability, infectivity and transmission. Since most data were obtained in traditional two-dimensional cell cultures, we will further attempt to place them into the context of polarized cultures and in vivo pathogenesis. Throughout the review, we highlight numerous knowledge gaps to stimulate future research into the egress mechanisms of these highly prevalent but largely understudied viruses.


Asunto(s)
Infecciones por Virus ARN/virología , Virus ARN/clasificación , Virión/fisiología , Liberación del Virus , Animales , Humanos , Virus ARN/fisiología
16.
Adv Virus Res ; 104: 97-122, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31439154

RESUMEN

Viruses must navigate the complex endomembranous network of the host cell to cause infection. In the case of a non-enveloped virus that lacks a surrounding lipid bilayer, endocytic uptake from the plasma membrane is not sufficient to cause infection. Instead, the virus must travel within organelle membranes to reach a specific cellular destination that supports exposure or arrival of the virus to the cytosol. This is achieved by viral penetration across a host endomembrane, ultimately enabling entry of the virus into the nucleus to initiate infection. In this review, we discuss the entry mechanisms of three distinct non-enveloped DNA viruses-adenovirus (AdV), human papillomavirus (HPV), and polyomavirus (PyV)-highlighting how each exploit different intracellular transport machineries and membrane penetration apparatus associated with the endosome, Golgi, and endoplasmic reticulum (ER) membrane systems to infect a host cell. These processes not only illuminate a highly-coordinated interplay between non-enveloped viruses and their host, but may provide new strategies to combat non-enveloped virus-induced diseases.


Asunto(s)
Adenoviridae/fisiología , Interacciones Huésped-Patógeno , Papillomaviridae/fisiología , Poliomavirus/fisiología , Internalización del Virus , Endocitosis , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Endosomas/metabolismo , Endosomas/virología , Aparato de Golgi/metabolismo , Aparato de Golgi/virología , Humanos
17.
Adv Exp Med Biol ; 1215: 45-68, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31317495

RESUMEN

To initiate infection, non-enveloped viruses must recognize a target cell and penetrate the cell membrane by pore formation or membrane lysis. Rotaviruses are non-enveloped dsRNA viruses that infect the mature intestinal epithelium. They are major etiologic agents of diarrheal disease in human infants, as well as in young individuals of various avian and mammalian species. Rotavirus entry into the cell is a complex multistep process initiated by the interaction of the tip of the viral spike with glycan ligands at the cell surface, and driven by conformational changes of the proteins present in the outer protein capsid, the viral machinery for entry. This review feeds on the abundant structural information produced for rotavirus during the past 30 years and focuses on the structure and the dynamics of the rotavirus entry machinery. We survey the current models for rotavirus entry into cells.


Asunto(s)
Infecciones por Rotavirus , Rotavirus , Internalización del Virus , Animales , Membrana Celular/virología , Humanos , Modelos Biológicos , Rotavirus/fisiología , Infecciones por Rotavirus/patología , Infecciones por Rotavirus/virología
18.
Adv Exp Med Biol ; 1215: 13-43, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31317494

RESUMEN

The most widely-used assays for studying viral entry, including infectivity, cofloatation, and cell-cell fusion assays, yield functional information but provide low resolution of individual entry steps. Structural characterization provides high-resolution conformational information, but on its own is unable to address the functional significance of these conformations. Single virion tracking microscopy techniques provide more detail on the intermediate entry steps than infection assays and more functional information than structural methods, bridging the gap between these methods. In addition, single virion approaches also provide dynamic information about the kinetics of entry processes. This chapter reviews single virion tracking techniques and describes how they can be applied to study specific virus entry steps. These techniques provide information complementary to traditional ensemble approaches. Single virion techniques may either probe virion behavior in live cells or in biomimetic platforms. Synthesizing information from ensemble, structural, and single virion techniques ultimately yields a more complete understanding of the viral entry process than can be achieved by any single method alone.


Asunto(s)
Biomimética , Células , Microscopía , Internalización del Virus , Células/virología , Virología/instrumentación
19.
J Mol Biol ; 431(17): 3124-3138, 2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31233764

RESUMEN

Rotaviruses, like other non-enveloped, double-strand RNA viruses, package an RNA-dependent RNA polymerase (RdRp) with each duplex of their segmented genomes. Rotavirus cell entry results in loss of an outer protein layer and delivery into the cytosol of an intact, inner capsid particle (the "double-layer particle," or DLP). The RdRp, designated VP1, is active inside the DLP; each VP1 achieves many rounds of mRNA transcription from its associated genome segment. Previous work has shown that one VP1 molecule lies close to each 5-fold axis of the icosahedrally symmetric DLP, just beneath the inner surface of its protein shell, embedded in tightly packed RNA. We have determined a high-resolution structure for the rotavirus VP1 RdRp in situ, by local reconstruction of density around individual 5-fold positions. We have analyzed intact virions ("triple-layer particles"), non-transcribing DLPs and transcribing DLPs. Outer layer dissociation enables the DLP to synthesize RNA, in vitro as well as in vivo, but appears not to induce any detectable structural change in the RdRp. Addition of NTPs, Mg2+, and S-adenosylmethionine, which allows active transcription, results in conformational rearrangements, in both VP1 and the DLP capsid shell protein, that allow a transcript to exit the polymerase and the particle. The position of VP1 (among the five symmetrically related alternatives) at one vertex does not correlate with its position at other vertices. This stochastic distribution of site occupancies limits long-range order in the 11-segment, double-strand RNA genome.


Asunto(s)
ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/metabolismo , Rotavirus/metabolismo , Sitios de Unión , Proteínas de la Cápside/química , Modelos Moleculares , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , ARN Bicatenario , Rotavirus/genética , Transcripción Genética , Proteínas del Núcleo Viral , Replicación Viral
20.
Cell Host Microbe ; 25(4): 617-629.e7, 2019 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-30926239

RESUMEN

The complement system is vital for anti-microbial defense. In the classical pathway, pathogen-bound antibody recruits the C1 complex (C1qC1r2C1s2) that initiates a cleavage cascade involving C2, C3, C4, and C5 and triggering microbial clearance. We demonstrate a C4-dependent antiviral mechanism that is independent of downstream complement components. C4 inhibits human adenovirus infection by directly inactivating the virus capsid. Rapid C4 activation and capsid deposition of cleaved C4b are catalyzed by antibodies via the classical pathway. Capsid-deposited C4b neutralizes infection independent of C2 and C3 but requires C1q antibody engagement. C4b inhibits capsid disassembly, preventing endosomal escape and cytosolic access. C4-deficient mice exhibit heightened viral burdens. Additionally, complement synergizes with the Fc receptor TRIM21 to block transduction by an adenovirus gene therapy vector but is partially restored by Fab virus shielding. These results suggest that the complement system could be altered to prevent virus infection and enhance virus gene therapy efficacy.


Asunto(s)
Infecciones por Adenovirus Humanos/inmunología , Adenovirus Humanos/inmunología , Cápside/metabolismo , Complemento C4/metabolismo , Inmunidad Humoral , Factores Inmunológicos/metabolismo , Inactivación de Virus , Animales , Anticuerpos Antivirales/metabolismo , Línea Celular , Complemento C1/metabolismo , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...