Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Cell Dev Biol ; 29: 551-69, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24099087

RESUMEN

Virus budding is a complex, multistep process in which viral proteins make specific alterations in membrane curvature. Many different viral proteins can deform the membrane and form a budding virion, but very few can mediate membrane scission to complete the budding process. As a result, enveloped viruses have developed numerous ways of facilitating membrane scission, including hijacking host cellular scission machinery and expressing their own scission proteins. These proteins mediate scission in very different ways, though the biophysical mechanics underlying their actions may be similar. In this review, we explore the mechanisms of membrane scission and the ways in which enveloped viruses use these systems to mediate the release of budding virions.


Asunto(s)
Liberación del Virus , Membrana Celular/química , Membrana Celular/virología , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Humanos , Proteínas Virales/metabolismo
2.
Cell ; 142(6): 902-13, 2010 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-20850012

RESUMEN

Many viruses utilize host ESCRT proteins for budding; however, influenza virus budding is thought to be ESCRT-independent. In this study we have found a role for the influenza virus M2 proton-selective ion channel protein in mediating virus budding. We observed that a highly conserved amphipathic helix located within the M2 cytoplasmic tail mediates a cholesterol-dependent alteration in membrane curvature. The 17 amino acid amphipathic helix is sufficient for budding into giant unilamellar vesicles, and mutation of this sequence inhibited budding of transfected M2 protein in vivo. We show that M2 localizes to the neck of budding virions and that mutation of the M2 amphipathic helix results in failure of the virus to undergo membrane scission and virion release. These data suggest that M2 mediates the final steps of budding for influenza viruses, bypassing the need for host ESCRT proteins.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Virus de la Influenza A/metabolismo , Proteínas de la Matriz Viral/metabolismo , Liberación del Virus , Animales , Línea Celular , Membrana Celular/química , Membrana Celular/metabolismo , Perros , Humanos , Virus de la Influenza A/ultraestructura , Lípidos de la Membrana/metabolismo , Microscopía Electrónica , Estructura Terciaria de Proteína , Proteínas de la Matriz Viral/análisis , Proteínas de la Matriz Viral/química
3.
Subcell Biochem ; 106: 441-459, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38159237

RESUMEN

The cholesterol of the host cell plasma membrane and viral M2 protein plays a crucial role in multiple stages of infection and replication of the influenza A virus. Cholesterol is required for the formation of heterogeneous membrane microdomains (or rafts) in the budozone of the host cell that serves as assembly sites for the viral components. The raft microstructures act as scaffolds for several proteins. Cholesterol may further contribute to the mechanical forces necessary for membrane scission in the last stage of budding and help to maintain the stability of the virus envelope. The M2 protein has been shown to cause membrane scission in model systems by promoting the formation of curved lipid bilayer structures that, in turn, can lead to membrane vesicles budding off or scission intermediates. Membrane remodeling by M2 is intimately linked with cholesterol as it affects local lipid composition, fluidity, and stability of the membrane. Thus, both cholesterol and M2 protein contribute to the efficient and proper release of newly formed influenza viruses from the virus-infected cells.


Asunto(s)
Virus de la Influenza A , Orthomyxoviridae , Virus de la Influenza A/metabolismo , Proteínas Virales/metabolismo , Colesterol/metabolismo , Microdominios de Membrana/metabolismo , Membrana Celular/metabolismo
4.
Emerg Infect Dis ; 29(2): 304-313, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36692336

RESUMEN

Lassa fever virus (LASV) is the causative agent of Lassa fever, a disease endemic in West Africa. Exploring the relationships between environmental factors and LASV transmission across ecologically diverse regions can provide crucial information for the design of appropriate interventions and disease monitoring. We investigated LASV exposure in 2 ecologically diverse regions of Guinea. Our results showed that exposure to LASV was heterogenous between and within sites. LASV IgG seropositivity was 11.9% (95% CI 9.7%-14.5%) in a coastal study site in Basse-Guinée, but it was 59.6% (95% CI 55.5%-63.5%) in a forested study site located in Guinée Forestière. Seropositivity increased with age in the coastal site. We also found significant associations between exposure risk for LASV and landscape fragmentation in coastal and forested regions. Our study highlights the potential link between environmental change and LASV emergence and the urgent need for research on land management practices that reduce disease risks.


Asunto(s)
Fiebre de Lassa , Humanos , Fiebre de Lassa/epidemiología , Guinea/epidemiología , Virus Lassa , África Occidental
5.
PLoS Pathog ; 17(6): e1009620, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34166465

RESUMEN

Questions persist as to the origin of the COVID-19 pandemic. Evidence is building that its origin as a zoonotic spillover occurred prior to the officially accepted timing of early December, 2019. Here we provide novel methods to date the origin of COVID-19 cases. We show that six countries had exceptionally early cases, unlikely to represent part of their main case series. The model suggests a likely timing of the first case of COVID-19 in China as November 17 (95% CI October 4). Origination dates are discussed for the first five countries outside China and each continent. Results infer that SARS-CoV-2 emerged in China in early October to mid-November, and by January, had spread globally. This suggests an earlier and more rapid timeline of spread. Our study provides new approaches for estimating dates of the arrival of infectious diseases based on small samples that can be applied to many epidemiological situations.


Asunto(s)
COVID-19 , Pandemias/historia , SARS-CoV-2 , Zoonosis , Animales , COVID-19/epidemiología , COVID-19/historia , COVID-19/transmisión , China/epidemiología , Historia del Siglo XXI , Humanos , Zoonosis/epidemiología , Zoonosis/historia , Zoonosis/transmisión
6.
Soft Matter ; 17(8): 2024-2027, 2021 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-33599656

RESUMEN

Lipid bilayer vesicles have provided a window into the function and fundamental properties of cells. However, as is the case for most living and soft matter, vesicles do not remain still. This necessitates some microscopy experiments to include a preparatory immobilisation step. Here, we describe a straightforward method to immobilise giant unilamellar vesicles (GUVs) using zirconium-based metal-organic frameworks (MOFs) and demonstrate that GUVs bound in this way will stay in position on a timescale of minutes to hours.


Asunto(s)
Estructuras Metalorgánicas , Liposomas Unilamelares , Membrana Dobles de Lípidos , Microscopía , Circonio
7.
Proc Natl Acad Sci U S A ; 115(37): E8595-E8603, 2018 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-30150411

RESUMEN

The influenza A matrix 2 (M2) transmembrane protein facilitates virion release from the infected host cell. In particular, M2 plays a role in the induction of membrane curvature and/or in the scission process whereby the envelope is cut upon virion release. Here we show using coarse-grained computer simulations that various M2 assembly geometries emerge due to an entropic driving force, resulting in compact clusters or linearly extended aggregates as a direct consequence of the lateral membrane stresses. Conditions under which these protein assemblies will cause the lipid membrane to curve are explored, and we predict that a critical cluster size is required for this to happen. We go on to demonstrate that under the stress conditions taking place in the cellular membrane as it undergoes large-scale membrane remodeling, the M2 protein will, in principle, be able to both contribute to curvature induction and sense curvature to line up in manifolds where local membrane line tension is high. M2 is found to exhibit linactant behavior in liquid-disordered-liquid-ordered phase-separated lipid mixtures and to be excluded from the liquid-ordered phase, in near-quantitative agreement with experimental observations. Our findings support a role for M2 in membrane remodeling during influenza viral budding both as an inducer and a sensor of membrane curvature, and they suggest a mechanism by which localization of M2 can occur as the virion assembles and releases from the host cell, independent of how the membrane curvature is produced.


Asunto(s)
Membrana Celular/virología , Proteínas de la Matriz Viral/fisiología , Ensamble de Virus , Liberación del Virus , Algoritmos , Animales , Membrana Celular/química , Membrana Celular/ultraestructura , Perros , Entropía , Interacciones Huésped-Patógeno , Células de Riñón Canino Madin Darby , Lípidos de la Membrana/química , Microscopía Electrónica , Modelos Biológicos , Simulación de Dinámica Molecular
8.
Bioinformatics ; 33(13): 1911-1915, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28200119

RESUMEN

MOTIVATION: Ebola viruses are not pathogenic but can be adapted to replicate and cause disease in rodents. Here, we used a structural bioinformatics approach to analyze the mutations associated with Ebola virus adaptation to rodents to elucidate the determinants of host-specific Ebola virus pathogenicity. RESULTS: We identified 33 different mutations associated with Ebola virus adaptation to rodents in the proteins GP, NP, L, VP24 and VP35. Only VP24, GP and NP were consistently found mutated in rodent-adapted Ebola virus strains. Fewer than five mutations in these genes seem to be required for the adaptation of Ebola viruses to a new species. The role of mutations in GP and NP is not clear. However, three VP24 mutations located in the protein interface with karyopherin α5 may enable VP24 to inhibit karyopherins and subsequently the host interferon response. Three further VP24 mutations change hydrogen bonding or cause conformational changes. Hence, there is evidence that few mutations including crucial mutations in VP24 enable Ebola virus adaptation to new hosts. Since Reston virus, the only non-human pathogenic Ebolavirus species circulates in pigs in Asia, this raises concerns that few mutations may result in novel human pathogenic Ebolaviruses. CONTACT: m.n.wass@kent.ac.uk , m.michaelis@kent.ac.uk or j.s.rossman@kent.ac.uk. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Ebolavirus/genética , Mutación , Roedores/virología , Proteínas Virales/genética , Animales , Cricetinae , Ebolavirus/metabolismo , Ebolavirus/patogenicidad , Evolución Molecular , Cobayas , Humanos , Ratones , Conformación Proteica , Proteínas Virales/metabolismo
9.
Biochemistry ; 55(25): 3493-6, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27299375

RESUMEN

Membrane scission is the final step in all budding processes wherein a membrane neck is sufficiently constricted so as to allow for fission and the release of the budded particle. For influenza viruses, membrane scission is mediated by an amphipathic helix (AH) domain in the viral M2 protein. While it is known that the M2AH alters membrane curvature, it is not known how the protein is localized to the center neck of budding virions where it would be able to cause membrane scission. Here, we use molecular dynamics simulations on buckled lipid bilayers to show that the M2AH senses membrane curvature and preferentially localizes to regions of high membrane curvature, comparable to that seen at the center neck of budding influenza viruses. These results were then validated using in vitro binding assays to show that the M2AH senses membrane curvature by detecting lipid packing defects in the membrane. Our results show that the M2AH senses membrane curvature and suggest that the AH domain may localize the protein at the viral neck where it can then mediate membrane scission and the release of budding viruses.


Asunto(s)
Membrana Celular/metabolismo , Virus de la Influenza A/metabolismo , Membrana Dobles de Lípidos/metabolismo , Lípidos de la Membrana/metabolismo , Proteínas de la Matriz Viral/metabolismo , Liberación del Virus/fisiología , Humanos
10.
Biochemistry ; 55(18): 2601-12, 2016 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-27089101

RESUMEN

Previous cell-penetrating peptides (CPPs) generally have low cytosolic delivery efficiencies, because of inefficient endosomal escape. In this study, a family of small, amphipathic cyclic peptides was found to be highly efficient CPPs, with cytosolic delivery efficiencies of up to 120% (compared to 2.0% for Tat). These cyclic CPPs bind directly to the plasma membrane phospholipids and enter mammalian cells via endocytosis, followed by efficient release from the endosome. Their total cellular uptake efficiency correlates positively with the binding affinity for the plasma membrane, whereas their endosomal escape efficiency increases with the endosomal membrane-binding affinity. The cyclic CPPs induce membrane curvature on giant unilamellar vesicles and budding of small vesicles, which subsequently collapse into amorphous lipid/peptide aggregates. These data suggest that cyclic CPPs exit the endosome by binding to the endosomal membrane and inducing CPP-enriched lipid domains to bud off as small vesicles. Together with their high proteolytic stability, low cytotoxicity, and oral bioavailability, these cyclic CPPs should provide a powerful system for intracellular delivery of therapeutic agents and chemical probes.


Asunto(s)
Péptidos de Penetración Celular , Endosomas/metabolismo , Membranas Intracelulares/metabolismo , Células A549 , Animales , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/farmacocinética , Péptidos de Penetración Celular/farmacología , Células HeLa , Humanos , Ratones , Células 3T3 NIH , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacocinética , Péptidos Cíclicos/farmacología , Permeabilidad
11.
J Virol ; 89(2): 1094-104, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25378494

RESUMEN

UNLABELLED: Transmembrane domains (TMDs) from single-spanning membrane proteins are commonly viewed as membrane anchors for functional domains. Influenza virus neuraminidase (NA) exemplifies this concept, as it retains enzymatic function upon proteolytic release from the membrane. However, the subtype 1 NA TMDs have become increasingly more polar in human strains since 1918, which suggests that selection pressure exists on this domain. Here, we investigated the N1 TMD-head domain relationship by exchanging a prototypical "old" TMD (1933) with a "recent" (2009), more polar TMD and an engineered hydrophobic TMD. Each exchange altered the TMD association, decreased the NA folding efficiency, and significantly reduced viral budding and replication at 37°C compared to at 33°C, at which NA folds more efficiently. Passaging the chimera viruses at 37°C restored the NA folding efficiency, viral budding, and infectivity by selecting for NA TMD mutations that correspond with their polar or hydrophobic assembly properties. These results demonstrate that single-spanning membrane protein TMDs can influence distal domain folding, as well as membrane-related processes, and suggest the NA TMD in H1N1 viruses has become more polar to maintain compatibility with the evolving enzymatic head domain. IMPORTANCE: The neuraminidase (NA) protein from influenza A viruses (IAVs) functions to promote viral release and is one of the major surface antigens. The receptor-destroying activity in NA resides in the distal head domain that is linked to the viral membrane by an N-terminal hydrophobic transmembrane domain (TMD). Over the last century, the subtype 1 NA TMDs (N1) in human H1N1 viruses have become increasingly more polar, and the head domains have changed to alter their antigenicity. Here, we provide the first evidence that an "old" N1 head domain from 1933 is incompatible with a "recent" (2009), more polar N1 TMD sequence and that, during viral replication, the head domain drives the selection of TMD mutations. These mutations modify the intrinsic TMD assembly to restore the head domain folding compatibility and the resultant budding deficiency. This likely explains why the N1 TMDs have become more polar and suggests the N1 TMD and head domain have coevolved.


Asunto(s)
Evolución Molecular , Subtipo H1N1 del Virus de la Influenza A/enzimología , Subtipo H1N1 del Virus de la Influenza A/fisiología , Neuraminidasa/genética , Neuraminidasa/metabolismo , Pliegue de Proteína , Estructura Terciaria de Proteína , Proteínas Virales/genética , Proteínas Virales/metabolismo , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Mutación , Temperatura , Liberación del Virus , Replicación Viral
12.
Biochem Soc Trans ; 44(4): 973-8, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27528741

RESUMEN

The ongoing Ebola virus (also known as Zaire ebolavirus, a member of the Ebolavirus family) outbreak in West Africa has so far resulted in >28000 confirmed cases compared with previous Ebolavirus outbreaks that affected a maximum of a few hundred individuals. Hence, Ebolaviruses impose a much greater threat than we may have expected (or hoped). An improved understanding of the virus biology is essential to develop therapeutic and preventive measures and to be better prepared for future outbreaks by members of the Ebolavirus family. Computational investigations can complement wet laboratory research for biosafety level 4 pathogens such as Ebolaviruses for which the wet experimental capacities are limited due to a small number of appropriate containment laboratories. During the current West Africa outbreak, sequence data from many Ebola virus genomes became available providing a rich resource for computational analysis. Here, we consider the studies that have already reported on the computational analysis of these data. A range of properties have been investigated including Ebolavirus evolution and pathogenicity, prediction of micro RNAs and identification of Ebolavirus specific signatures. However, the accuracy of the results remains to be confirmed by wet laboratory experiments. Therefore, communication and exchange between computational and wet laboratory researchers is necessary to make maximum use of computational analyses and to iteratively improve these approaches.


Asunto(s)
Biología Computacional/métodos , Brotes de Enfermedades/prevención & control , Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/epidemiología , Fiebre Hemorrágica Ebola/virología , África Occidental/epidemiología , Ebolavirus/clasificación , Ebolavirus/genética , Evolución Molecular , Genoma Viral/genética , Genómica/métodos , Humanos , Proteínas Virales/genética
13.
Biochem Soc Trans ; 42(5): 1425-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25233426

RESUMEN

Influenza A virus belongs to the Orthomyxoviridae family. It is an enveloped virus that contains a segmented and negative-sense RNA genome. Influenza A viruses cause annual epidemics and occasional major pandemics, are a major cause of morbidity and mortality worldwide, and have a significant financial impact on society. Assembly and budding of new viral particles are a complex and multi-step process involving several host and viral factors. Influenza viruses use lipid raft domains in the apical plasma membrane of polarized epithelial cells as sites of budding. Two viral glycoproteins, haemagglutinin and neuraminidase, concentrate in lipid rafts, causing alterations in membrane curvature and initiation of the budding process. Matrix protein 1 (M1), which forms the inner structure of the virion, is then recruited to the site followed by incorporation of the viral ribonucleoproteins and matrix protein 2 (M2). M1 can alter membrane curvature and progress budding, whereas lipid raft-associated M2 stabilizes the site of budding, allowing for proper assembly of the virion. In the later stages of budding, M2 is localized to the neck of the budding virion at the lipid phase boundary, where it causes negative membrane curvature, leading to scission and virion release.


Asunto(s)
Membrana Celular/química , Células Epiteliales/virología , Virus de la Influenza A/fisiología , Liberación del Virus , Membrana Celular/metabolismo , Membrana Celular/virología , Polaridad Celular , Forma de la Célula , Células Epiteliales/química , Células Epiteliales/metabolismo , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Humanos , Microdominios de Membrana , Neuraminidasa/química , Neuraminidasa/metabolismo , Transporte de Proteínas , Proteínas Virales/química , Proteínas Virales/metabolismo
14.
Nat Commun ; 15(1): 4171, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755147

RESUMEN

Human Ebola virus (EBOV) outbreaks caused by persistent EBOV infection raises questions on the role of zoonotic spillover in filovirus epidemiology. To characterise filovirus zoonotic exposure, we collected cross-sectional serum samples from bushmeat hunters (n = 498) in Macenta Prefecture Guinea, adjacent to the index site of the 2013 EBOV-Makona spillover event. We identified distinct immune signatures (20/498, 4.0%) to multiple EBOV antigens (GP, NP, VP40) using stepwise ELISA and Western blot analysis and, live EBOV neutralisation (5/20; 25%). Using comparative serological data from PCR-confirmed survivors of the 2013-2016 EBOV outbreak, we demonstrated that most signatures (15/20) were not plausibly explained by prior EBOV-Makona exposure. Subsequent data-driven modelling of EBOV immunological outcomes to remote-sensing environmental data also revealed consistent associations with intact closed canopy forest. Together our findings suggest exposure to other closely related filoviruses prior to the 2013-2016 West Africa epidemic and highlight future surveillance priorities.


Asunto(s)
Anticuerpos Antivirales , Ebolavirus , Fiebre Hemorrágica Ebola , Humanos , Animales , Guinea/epidemiología , Ebolavirus/inmunología , Ebolavirus/aislamiento & purificación , Fiebre Hemorrágica Ebola/epidemiología , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/virología , Fiebre Hemorrágica Ebola/sangre , Fiebre Hemorrágica Ebola/transmisión , Adulto , Masculino , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Persona de Mediana Edad , Zoonosis/virología , Zoonosis/epidemiología , Zoonosis/transmisión , Femenino , Estudios Transversales , Brotes de Enfermedades , Adulto Joven , Anciano , Ensayo de Inmunoadsorción Enzimática , Zoonosis Virales/epidemiología , Zoonosis Virales/transmisión , Zoonosis Virales/virología , Antígenos Virales/inmunología
15.
J Virol ; 86(20): 10950-60, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22875971

RESUMEN

Influenza virus is pleiomorphic, producing both spherical (100-nm-diameter) and filamentous (100-nm by 20-µm) virions. While the spherical virions are known to enter host cells through exploitation of clathrin-mediated endocytosis, the entry pathway for filamentous virions has not been determined, though the existence of an alternative, non-clathrin-, non-caveolin-mediated entry pathway for influenza virus has been known for many years. In this study, we confirm recent results showing that influenza virus utilizes macropinocytosis as an alternate entry pathway. Furthermore, we find that filamentous influenza viruses use macropinocytosis as the primary entry mechanism. Virions enter cells as intact filaments within macropinosomes and are trafficked to the acidic late-endosomal compartment. Low pH triggers a conformational change in the M2 ion channel protein, altering membrane curvature and leading to a fragmentation of the filamentous virions. This fragmentation may enable more-efficient fusion between the viral and endosomal membranes.


Asunto(s)
Virus de la Hepatitis A/fisiología , Pinocitosis , Proteínas de la Matriz Viral/metabolismo , Internalización del Virus , Animales , Línea Celular , Perros , Endocitosis , Endosomas/metabolismo , Endosomas/virología , Células HEK293 , Humanos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Microscopía Electrónica , Conformación Proteica , Proteínas Virales de Fusión , Proteínas de la Matriz Viral/química
16.
Front Immunol ; 14: 1150105, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37122723

RESUMEN

The mechanism of Long Covid (Post-Acute Sequelae of COVID-19; PASC) is currently unknown, with no validated diagnostics or therapeutics. SARS-CoV-2 can cause disseminated infections that result in multi-system tissue damage, dysregulated inflammation, and cellular metabolic disruptions. The tissue damage and inflammation has been shown to impair microvascular circulation, resulting in hypoxia, which coupled with virally-induced metabolic reprogramming, increases cellular anaerobic respiration. Both acute and PASC patients show systemic dysregulation of multiple markers of the acid-base balance. Based on these data, we hypothesize that the shift to anaerobic respiration causes an acid-base disruption that can affect every organ system and underpins the symptoms of PASC. This hypothesis can be tested by longitudinally evaluating acid-base markers in PASC patients and controls over the course of a month. If our hypothesis is correct, this could have significant implications for our understanding of PASC and our ability to develop effective diagnostic and therapeutic approaches.


Asunto(s)
COVID-19 , Síndrome Post Agudo de COVID-19 , Humanos , SARS-CoV-2 , Equilibrio Ácido-Base , Inflamación
17.
Vaccines (Basel) ; 10(5)2022 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-35632408

RESUMEN

Long COVID is a multi-system syndrome following SARS-CoV-2 infection with persistent symptoms of at least 4 weeks, and frequently for several months. It has been suggested that there may be an autoimmune component. There has been an understandable caution amongst some people experiencing long COVID that, by boosting their immune response, a COVID vaccine may exacerbate their symptoms. We aimed to survey people living with long COVID, evaluating the impact of their first COVID vaccination on their symptoms. METHODS: Patients with long COVID were invited to complete a web-based questionnaire through postings on social media and direct mailing from support groups. Basic demographics, range and severity of long COVID symptoms, before and after their vaccine, were surveyed. RESULTS: 900 people participated in the questionnaire, of whom 45 had pre-existing myalgic encephalomyelitis or chronic fatigue syndrome (ME/CFS) but no evidence of COVID infection, and a further 43 did not complete the survey in full. The demographics and symptomology of the remaining 812 people were similar to those recorded by the UK Office of National Statistics. Following vaccination, 57.9% of participants reported improvements in symptoms, 17.9% reported deterioration and the remainder no change. There was considerable individual variation in responses. Larger improvements in symptom severity scores were seen in those receiving the mRNA vaccines compared to adenoviral vector vaccines. CONCLUSIONS: Our survey suggests COVID-19 vaccination may improve long COVID patients, on average. The observational nature of the survey limits drawing direct causal inference, but requires validation with a randomised controlled trial.

18.
J Virol ; 84(10): 5078-88, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20219914

RESUMEN

Influenza A virus buds from cells as spherical (approximately 100-nm diameter) and filamentous (approximately 100 nm x 2 to 20 microm) virions. Previous work has determined that the matrix protein (M1) confers the ability of the virus to form filaments; however, additional work has suggested that the influenza virus M2 integral membrane protein also plays a role in viral filament formation. In examining the role of the M2 protein in filament formation, we observed that the cytoplasmic tail of M2 contains several sites that are essential for filament formation. Additionally, whereas M2 is a nonraft protein, expression of other viral proteins in the context of influenza virus infection leads to the colocalization of M2 with sites of virus budding and lipid raft domains. We found that an amphipathic helix located within the M2 cytoplasmic tail is able to bind cholesterol, and we speculate that M2 cholesterol binding is essential for both filament formation and the stability of existing viral filaments.


Asunto(s)
Virus de la Influenza A/fisiología , Proteínas de la Matriz Viral/fisiología , Virión/ultraestructura , Ensamble de Virus , Secuencia de Aminoácidos , Animales , Línea Celular , Colesterol/metabolismo , Perros , Humanos , Microscopía Electrónica , Microscopía Fluorescente , Datos de Secuencia Molecular , Unión Proteica
19.
J Phys Chem B ; 124(31): 6738-6747, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32644803

RESUMEN

The influenza virus M2 amphipathic helix (M2AH) alters membrane curvature in a cholesterol-dependent manner, mediating viral membrane scission during influenza virus budding. Here, we have investigated the biophysical effects of cholesterol on the ability of an M2AH peptide to manipulate membrane properties. We see that the ability of the M2AH to interact with membranes and form an α-helix is independent of membrane cholesterol concentration; however, cholesterol affects the angle of the M2AH peptide within the membrane. This change in membrane orientation affects the ability of the M2AH to alter lipid order. In low-cholesterol membranes, the M2AH is inserted near the level of the lipid head groups, increasing lipid order, which may contribute to generation of the membrane curvature. As the cholesterol content increases, the M2AH insertion becomes flatter and slightly deeper in the membrane below the lipid headgroups, where the polar face can continue to interact with the headgroups while the hydrophobic face binds cholesterol. This changed orientation minimizes lipid packing defects and lipid order changes, likely reducing the generation of membrane curvature. Thus, cholesterol regulates M2 membrane scission by precisely modulating M2AH positioning within the membrane. This has implications for the understanding of many of amphipathic-helix-driven cellular budding processes that occur in specific lipid environments.


Asunto(s)
Orthomyxoviridae , Proteínas de la Matriz Viral , Membrana Celular , Colesterol , Membrana Dobles de Lípidos , Liberación del Virus
20.
Mol Biol Cell ; 17(5): 2166-76, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16495340

RESUMEN

Stimulation of the T-cell receptor (TCR) results in the activation of several transcription factors, including NF-kappaB, that are crucial for T-cell proliferation and gain of effector functions. On TCR engagement, several proteins within the TCR-directed NF-kappaB signaling pathway undergo dynamic spatial redistribution, but the significance of these redistribution events is largely unknown. We have previously described TCR-induced cytoplasmic structures called POLKADOTS (punctate and oligomeric killing or activating domains transducing signals) that are enriched in the NF-kappaB signaling intermediate, Bcl10. We now show that these structures are formed only under conditions that promote efficient NF-kappaB activation. Furthermore, POLKADOTS formation is dependent on functional domains of specific NF-kappaB signal transducers. Through use of a photoactivatable GFP, we demonstrate that POLKADOTS contain both a highly stable and a rapidly equilibrating protein component. FRET analyses show that POLKADOTS are sites of enriched interactions between Bcl10 and partner signaling proteins. These observations strongly suggest that POLKADOTS are focal sites of dynamic information exchange between cytosolic intermediates in the process of TCR activation of NF-kappaB.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , FN-kappa B/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/ultraestructura , Proteínas Adaptadoras Transductoras de Señales/análisis , Animales , Proteína 10 de la LLC-Linfoma de Células B , Caspasas/análisis , Caspasas/metabolismo , Células Cultivadas , Humanos , Ratones , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas , FN-kappa B/agonistas , Proteínas de Neoplasias/análisis , Proteínas de Neoplasias/metabolismo , Receptores de Antígenos de Linfocitos T/análisis , Transducción de Señal , Linfocitos T/química , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA