Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.966
Filtrar
1.
J Gen Virol ; 105(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38695734

RESUMEN

Members of the family Fimoviridae are plant viruses with a multipartite negative-sense enveloped RNA genome (-ssRNA), composed of 4-10 segments comprising 12.3-18.5 kb in total, within quasi-spherical virions. Fimoviruses are transmitted to plants by eriophyid mites and induce characteristic cytopathologies in their host plants, including double membrane-bound bodies in the cytoplasm of virus-infected cells. Most fimoviruses infect dicotyledonous plants, and many cause serious disease epidemics. This is a summary of the ICTV Report on the family Fimoviridae, which is available at ictv.global/report/fimoviridae.


Asunto(s)
Genoma Viral , Enfermedades de las Plantas , Virus de Plantas , Enfermedades de las Plantas/virología , Animales , Virus de Plantas/genética , Virus de Plantas/clasificación , Virus de Plantas/fisiología , ARN Viral/genética , Virión/ultraestructura , Plantas/virología , Virus ARN de Sentido Negativo/genética , Virus ARN de Sentido Negativo/clasificación , Ácaros/virología , Filogenia
2.
J Gen Virol ; 105(4)2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38687001

RESUMEN

Nairoviridae is a family for negative-sense RNA viruses with genomes of about 17.2-21.1 kb. These viruses are maintained in and/or transmitted by arthropods among birds, reptiles and mammals. Norwaviruses and orthonairoviruses can cause febrile illness in humans. Several orthonairoviruses can infect mammals, causing mild, severe and sometimes, fatal diseases. Nairovirids produce enveloped virions containing two or three single-stranded RNA segments with open reading frames that encode a nucleoprotein (N), sometimes a glycoprotein precursor (GPC), and a large (L) protein containing an RNA-directed RNA polymerase (RdRP) domain. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) report on the family Nairoviridae, which is available at www.ictv.global/report/nairoviridae.


Asunto(s)
Genoma Viral , Animales , Humanos , Sistemas de Lectura Abierta , Proteínas Virales/genética , Nairovirus/genética , Nairovirus/clasificación , Nairovirus/aislamiento & purificación , ARN Viral/genética , Filogenia , Virión/ultraestructura , ARN Polimerasa Dependiente del ARN/genética
3.
J Virol ; 98(5): e0006824, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38661364

RESUMEN

The portal protein of tailed bacteriophage plays essential roles in various aspects of capsid assembly, motor assembly, genome packaging, connector formation, and infection processes. After DNA packaging is complete, additional proteins are assembled onto the portal to form the connector complex, which is crucial as it bridges the mature head and tail. In this study, we report high-resolution cryo-electron microscopy (cryo-EM) structures of the portal vertex from bacteriophage lambda in both its prohead and mature virion states. Comparison of these structures shows that during head maturation, in addition to capsid expansion, the portal protein undergoes conformational changes to establish interactions with the connector proteins. Additionally, the independently assembled tail undergoes morphological alterations at its proximal end, facilitating its connection to the head-tail joining protein and resulting in the formation of a stable portal-connector-tail complex. The B-DNA molecule spirally glides through the tube, interacting with the nozzle blade region of the middle-ring connector protein. These insights elucidate a mechanism for portal maturation and DNA translocation within the phage lambda system. IMPORTANCE: The tailed bacteriophages possess a distinct portal vertex that consists of a ring of 12 portal proteins associated with a 5-fold capsid shell. This portal protein is crucial in multiple stages of virus assembly and infection. Our research focused on examining the structures of the portal vertex in both its preliminary prohead state and the fully mature virion state of bacteriophage lambda. By analyzing these structures, we were able to understand how the portal protein undergoes conformational changes during maturation, the mechanism by which it prevents DNA from escaping, and the process of DNA spirally gliding.


Asunto(s)
Bacteriófago lambda , Proteínas de la Cápside , Cápside , Microscopía por Crioelectrón , Virión , Ensamble de Virus , Bacteriófago lambda/fisiología , Bacteriófago lambda/genética , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/química , Virión/metabolismo , Virión/ultraestructura , Cápside/metabolismo , Cápside/ultraestructura , ADN Viral/genética , ADN Viral/metabolismo , Empaquetamiento del ADN , Modelos Moleculares , Conformación Proteica
4.
J Med Virol ; 96(4): e29620, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38647027

RESUMEN

Vertical transmission has been described following monkeypox virus (MPXV) infection in pregnant women. The presence of MPXV has been reported in the placenta from infected women, but whether pathogens colonize placenta remains unexplored. We identify trophoblasts as a target cell for MPXV replication. In a pan-microscopy approach, we decipher the specific infectious cycle of MPXV and inner cellular structures in trophoblasts. We identified the formation of a specialized region for viral morphogenesis and replication in placental cells. We also reported infection-induced cellular remodeling. We found that MPXV stimulates cytoskeleton reorganization with intercellular extensions for MPXV cell spreading specifically to trophoblastic cells. Altogether, the specific infectious cycle of MPXV in trophoblast cells and these protrusions that were structurally and morphologically similar to filopodia reveal new insights into the infection of MPXV.


Asunto(s)
Monkeypox virus , Seudópodos , Trofoblastos , Trofoblastos/virología , Humanos , Seudópodos/virología , Femenino , Embarazo , Monkeypox virus/fisiología , Liberación del Virus , Replicación Viral , Citoesqueleto/virología , Placenta/virología , Placenta/citología , Virión/ultraestructura , Microscopía/métodos , Línea Celular
5.
Cell ; 187(9): 2236-2249.e17, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38614100

RESUMEN

Unlike those of double-stranded DNA (dsDNA), single-stranded DNA (ssDNA), and ssRNA viruses, the mechanism of genome packaging of dsRNA viruses is poorly understood. Here, we combined the techniques of high-resolution cryoelectron microscopy (cryo-EM), cellular cryoelectron tomography (cryo-ET), and structure-guided mutagenesis to investigate genome packaging and capsid assembly of bluetongue virus (BTV), a member of the Reoviridae family of dsRNA viruses. A total of eleven assembly states of BTV capsid were captured, with resolutions up to 2.8 Å, with most visualized in the host cytoplasm. ATPase VP6 was found underneath the vertices of capsid shell protein VP3 as an RNA-harboring pentamer, facilitating RNA packaging. RNA packaging expands the VP3 shell, which then engages middle- and outer-layer proteins to generate infectious virions. These revealed "duality" characteristics of the BTV assembly mechanism reconcile previous contradictory co-assembly and core-filling models and provide insights into the mysterious RNA packaging and capsid assembly of Reoviridae members and beyond.


Asunto(s)
Virus de la Lengua Azul , Proteínas de la Cápside , Cápside , Microscopía por Crioelectrón , ARN Viral , Empaquetamiento del Genoma Viral , Virus de la Lengua Azul/genética , Virus de la Lengua Azul/fisiología , Virus de la Lengua Azul/metabolismo , Cápside/metabolismo , Cápside/ultraestructura , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/química , Animales , ARN Viral/metabolismo , ARN Viral/genética , Genoma Viral/genética , Ensamble de Virus , Tomografía con Microscopio Electrónico , Virión/metabolismo , Virión/genética , Virión/ultraestructura , Modelos Moleculares , Línea Celular , Cricetinae
6.
J Virol ; 98(3): e0153623, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38315014

RESUMEN

African swine fever (ASF) is a highly contagious viral disease that affects domestic and wild pigs. The causative agent of ASF is African swine fever virus (ASFV), a large double-stranded DNA virus with a complex virion structure. Among the various proteins encoded by ASFV, A137R is a crucial structural protein associated with its virulence. However, the structure and molecular mechanisms underlying the functions of A137R remain largely unknown. In this study, we present the structure of A137R determined by cryogenic electron microscopy single-particle reconstruction, which reveals that A137R self-oligomerizes to form a dodecahedron-shaped cage composed of 60 polymers. The dodecahedron is literally equivalent to a T = 1 icosahedron where the icosahedral vertexes are located in the center of each dodecahedral facet. Within each facet, five A137R protomers are arranged in a head-to-tail orientation with a long N-terminal helix forming the edge through which adjacent facets stitch together to form the dodecahedral cage. Combining structural analysis and biochemical evidence, we demonstrate that the N-terminal domain of A137R is crucial and sufficient for mediating the assembly of the dodecahedron. These findings imply the role of A137R cage as a core component in the icosahedral ASFV virion and suggest a promising molecular scaffold for nanotechnology applications. IMPORTANCE: African swine fever (ASF) is a lethal viral disease of pigs caused by African swine fever virus (ASFV). No commercial vaccines and antiviral treatments are available for the prevention and control of the disease. A137R is a structural protein of ASFV that is associated with its virulence. The discovery of the dodecahedron-shaped cage structure of A137R in this study is of great importance in understanding ASFV pathogenicity. This finding sheds light on the molecular mechanisms underlying the functions of A137R. Furthermore, the dodecahedral cage formed by A137R shows promise as a molecular scaffold for nanoparticle vectors. Overall, this study provides valuable insights into the structure and function of A137R, contributing to our understanding of ASFV and potentially opening up new avenues for the development of vaccines or treatments for ASF.


Asunto(s)
Virus de la Fiebre Porcina Africana , Porcinos , Proteínas Estructurales Virales , Animales , Fiebre Porcina Africana/virología , Virus de la Fiebre Porcina Africana/química , Virus de la Fiebre Porcina Africana/crecimiento & desarrollo , Virus de la Fiebre Porcina Africana/patogenicidad , Virus de la Fiebre Porcina Africana/ultraestructura , Microscopía por Crioelectrón , Relación Estructura-Actividad , Porcinos/virología , Proteínas Estructurales Virales/química , Proteínas Estructurales Virales/metabolismo , Proteínas Estructurales Virales/ultraestructura , Virión/química , Virión/metabolismo , Virión/ultraestructura , Virulencia
7.
Nature ; 623(7989): 1026-1033, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37993716

RESUMEN

Human immunodeficiency virus 1 (HIV-1) infection is initiated by binding of the viral envelope glycoprotein (Env) to the cell-surface receptor CD41-4. Although high-resolution structures of Env in a complex with the soluble domains of CD4 have been determined, the binding process is less understood in native membranes5-13. Here we used cryo-electron tomography to monitor Env-CD4 interactions at the membrane-membrane interfaces formed between HIV-1 and CD4-presenting virus-like particles. Env-CD4 complexes organized into clusters and rings, bringing the opposing membranes closer together. Env-CD4 clustering was dependent on capsid maturation. Subtomogram averaging and classification revealed that Env bound to one, two and finally three CD4 molecules, after which Env adopted an open state. Our data indicate that asymmetric HIV-1 Env trimers bound to one and two CD4 molecules are detectable intermediates during virus binding to host cell membranes, which probably has consequences for antibody-mediated immune responses and vaccine immunogen design.


Asunto(s)
Antígenos CD4 , Membrana Celular , Proteína gp120 de Envoltorio del VIH , VIH-1 , Multimerización de Proteína , Humanos , Vacunas contra el SIDA/química , Vacunas contra el SIDA/inmunología , Cápside/química , Cápside/metabolismo , Cápside/ultraestructura , Antígenos CD4/química , Antígenos CD4/metabolismo , Antígenos CD4/ultraestructura , Membrana Celular/química , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Microscopía por Crioelectrón , Tomografía con Microscopio Electrónico , Anticuerpos Anti-VIH/inmunología , Proteína gp120 de Envoltorio del VIH/química , Proteína gp120 de Envoltorio del VIH/metabolismo , Proteína gp120 de Envoltorio del VIH/ultraestructura , Infecciones por VIH/virología , VIH-1/química , VIH-1/ultraestructura , Virión/química , Virión/metabolismo , Virión/ultraestructura
8.
Phys Chem Chem Phys ; 25(18): 12882-12890, 2023 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-37165855

RESUMEN

The global spread of the new coronavirus COVID-19 has seriously affected human health and has caused a large number of deaths. Using molecular dynamics (MD) simulations to study the microscopic dynamic behavior of the virion provides an important means to study the pathogenic mechanism. In this work, we develop an ultra-coarse-grained (UCG) model of the SARS-CoV-2 virion from the authentic cryo-electron microscopy data, which enables MD simulation of the entire virion within microseconds. In addition, a hybrid all-atom and UCG (AA/UCG) virion model involving an all-atom spike protein is developed for the investigation of the spike protein interactions. A comparison of the conformational changes for the spike proteins as simulated in the hybrid model and that isolated in solution as in the free form reveals that the former is completely different from the latter. The simulation results demonstrate the necessity for the development of multiscale models to study the functions of proteins in the biomolecular complexes.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/metabolismo , Microscopía por Crioelectrón , Glicoproteína de la Espiga del Coronavirus/metabolismo , Simulación de Dinámica Molecular , Virión/metabolismo , Virión/ultraestructura
9.
Nature ; 617(7960): 409-416, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37138077

RESUMEN

CrAssphage and related viruses of the order Crassvirales (hereafter referred to as crassviruses) were originally discovered by cross-assembly of metagenomic sequences. They are the most abundant viruses in the human gut, are found in the majority of individual gut viromes, and account for up to 95% of the viral sequences in some individuals1-4. Crassviruses are likely to have major roles in shaping the composition and functionality of the human microbiome, but the structures and roles of most of the virally encoded proteins are unknown, with only generic predictions resulting from bioinformatic analyses4,5. Here we present a cryo-electron microscopy reconstruction of Bacteroides intestinalis virus ΦcrAss0016, providing the structural basis for the functional assignment of most of its virion proteins. The muzzle protein forms an assembly about 1 MDa in size at the end of the tail and exhibits a previously unknown fold that we designate the 'crass fold', that is likely to serve as a gatekeeper that controls the ejection of cargos. In addition to packing the approximately 103 kb of virus DNA, the ΦcrAss001 virion has extensive storage space for virally encoded cargo proteins in the capsid and, unusually, within the tail. One of the cargo proteins is present in both the capsid and the tail, suggesting a general mechanism for protein ejection, which involves partial unfolding of proteins during their extrusion through the tail. These findings provide a structural basis for understanding the mechanisms of assembly and infection of these highly abundant crassviruses.


Asunto(s)
Virus ADN , Intestinos , Proteínas Virales , Virión , Humanos , Cápside/química , Cápside/metabolismo , Cápside/ultraestructura , Microscopía por Crioelectrón , Virus ADN/química , Virus ADN/clasificación , Virus ADN/aislamiento & purificación , Virus ADN/metabolismo , Virus ADN/ultraestructura , Virión/química , Virión/metabolismo , Virión/ultraestructura , Ensamble de Virus , Intestinos/microbiología , Intestinos/virología , Proteínas Virales/química , Proteínas Virales/metabolismo , Proteínas Virales/ultraestructura , Desplegamiento Proteico , Pliegue de Proteína
10.
Viruses ; 14(12)2022 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-36560790

RESUMEN

Infection with the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative agent of the COVID-19 pandemic, leads to profound remodeling of cellular membranes, promoting viral replication and virion assembly. A full understanding of this drastic remodeling and the process of virion morphogenesis remains lacking. In this study, we applied room temperature transmission electron microscopy (TEM) and scanning transmission electron microscopy (STEM) tomography to visualize the SARS-CoV-2 replication factory in Vero cells, and present our results in comparison with published cryo-EM studies. We obtained cryo-EM-like clarity of the ultrastructure by employing high-pressure freezing, freeze substitution (HPF-FS) and embedding, allowing room temperature visualization of double-membrane vesicles (DMVs) in a near-native state. In addition, our data illustrate the consecutive stages of virion morphogenesis and reveal that SARS-CoV-2 ribonucleoprotein assembly and membrane curvature occur simultaneously. Finally, we show the tethering of virions to the plasma membrane in 3D, and that accumulations of virus particles lacking spike protein in large vesicles are most likely not a result of defective virion assembly at their membrane. In conclusion, this study puts forward a room-temperature EM technique providing near-native ultrastructural information about SARS-CoV-2 replication, adding to our understanding of the interaction of this pandemic virus with its host cell.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Chlorocebus aethiops , Humanos , Células Vero , Pandemias , Virión/ultraestructura
11.
J Virol ; 96(24): e0136722, 2022 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-36448797

RESUMEN

Coxsackievirus A9 (CVA9), an enterovirus, is a common cause of pediatric aseptic meningitis and neonatal sepsis. During cell entry, enterovirus capsids undergo conformational changes leading to expansion, formation of large pores, externalization of VP1 N termini, and loss of the lipid factor from VP1. Factors such as receptor binding, heat, and acidic pH can trigger capsid expansion in some enteroviruses. Here, we show that fatty acid-free bovine serum albumin or neutral endosomal ionic conditions can independently prime CVA9 for expansion and genome release. Our results showed that CVA9 treatment with albumin or endosomal ions generated a heterogeneous population of virions, which could be physically separated by asymmetric flow field flow fractionation and computationally by cryo-electron microscopy (cryo-EM) and image processing. We report cryo-EM structures of CVA9 A-particles obtained by albumin or endosomal ion treatment and a control nonexpanded virion to 3.5, 3.3, and 2.9 Å resolution, respectively. Whereas albumin promoted stable expanded virions, the endosomal ionic concentrations induced unstable CVA9 virions which easily disintegrated, losing their genome. Loss of most of the VP4 molecules and exposure of negatively charged amino acid residues in the capsid's interior after expansion created a repulsive viral RNA-capsid interface, aiding genome release. IMPORTANCE Coxsackievirus A9 (CVA9) is a common cause of meningitis and neonatal sepsis. The triggers and mode of action of RNA release into the cell unusually do not require receptor interaction. Rather, a slow process in the endosome, independent of low pH, is required. Here, we show by biophysical separation, cryogenic electron microscopy, and image reconstruction that albumin and buffers mimicking the endosomal ion composition can separately and together expand and prime CVA9 for uncoating. Furthermore, we show in these expanded particles that VP4 is present at only ~10% of the occupancy found in the virion, VP1 is externalized, and the genome is repelled by the negatively charged, repulsive inner surface of the capsid that occurs due to the expansion. Thus, we can now link observations from cell biology of infection with the physical processes that occur in the capsid to promote genome uncoating.


Asunto(s)
Cationes , Enterovirus Humano B , Humanos , Albúminas/farmacología , Proteínas de la Cápside/metabolismo , Cationes/farmacología , Microscopía por Crioelectrón , Endosomas/metabolismo , Enterovirus Humano B/efectos de los fármacos , Enterovirus Humano B/genética , Enterovirus Humano B/ultraestructura , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/virología , ARN/metabolismo , Virión/efectos de los fármacos , Virión/metabolismo , Virión/ultraestructura , Genoma Viral
12.
J Virol ; 96(16): e0062722, 2022 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-35924923

RESUMEN

Rotavirus live-attenuated vaccines, both mono- and pentavalent, generate broadly heterotypic protection. B-cells isolated from adults encode neutralizing antibodies, some with affinity for VP5*, that afford broad protection in mice. We have mapped the epitope of one such antibody by determining the high-resolution cryo-EM structure of its antigen-binding fragment (Fab) bound to the virion of a candidate vaccine strain, CDC-9. The Fab contacts both the distal end of a VP5* ß-barrel domain and the two VP8* lectin-like domains at the tip of a projecting spike. Its interactions with VP8* do not impinge on the likely receptor-binding site, suggesting that the mechanism of neutralization is at a step subsequent to initial attachment. We also examined structures of CDC-9 virions from two different stages of serial passaging. Nearly all the VP4 (cleaved to VP8*/VP5*) spikes on particles from the earlier passage (wild-type isolate) had transitioned from the "upright" conformation present on fully infectious virions to the "reversed" conformation that is probably the end state of membrane insertion, unable to mediate penetration, consistent with the very low in vitro infectivity of the wild-type isolate. About half the VP4 spikes were upright on particles from the later passage, which had recovered substantial in vitro infectivity but had acquired an attenuated phenotype in neonatal rats. A mutation in VP4 that occurred during passaging appears to stabilize the interface at the apex of the spike and could account for the greater stability of the upright spikes on the late-passage, attenuated isolate. IMPORTANCE Rotavirus live-attenuated vaccines generate broadly heterotypic protection, and B-cells isolated from adults encode antibodies that are broadly protective in mice. Determining the structural and mechanistic basis of broad protection can contribute to understanding the current limitations of vaccine efficacy in developing countries. The structure of an attenuated human rotavirus isolate (CDC-9) bound with the Fab fragment of a broadly heterotypic protective antibody shows that protection is probably due to inhibition of the conformational transition in the viral spike protein (VP4) critical for viral penetration, rather than to inhibition of receptor binding. A comparison of structures of CDC-9 virus particles at two stages of serial passaging supports a proposed mechanism for initial steps in rotavirus membrane penetration.


Asunto(s)
Anticuerpos ampliamente neutralizantes , Proteínas de la Cápside , Epítopos de Linfocito B , Rotavirus , Vacunas Atenuadas , Virión , Animales , Anticuerpos ampliamente neutralizantes/inmunología , Anticuerpos ampliamente neutralizantes/ultraestructura , Proteínas de la Cápside/química , Proteínas de la Cápside/inmunología , Proteínas de la Cápside/ultraestructura , Microscopía por Crioelectrón , Epítopos de Linfocito B/inmunología , Epítopos de Linfocito B/ultraestructura , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fab de Inmunoglobulinas/ultraestructura , Ratones , Conformación Proteica , Ratas , Rotavirus/química , Rotavirus/clasificación , Rotavirus/inmunología , Rotavirus/fisiología , Pase Seriado , Vacunas Atenuadas/química , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/metabolismo , Virión/inmunología , Virión/metabolismo , Virión/ultraestructura
13.
J Virol ; 96(17): e0060422, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-35939401

RESUMEN

Enterovirus 70 (EV70) is a human pathogen belonging to the family Picornaviridae. EV70 is transmitted by eye secretions and causes acute hemorrhagic conjunctivitis, a serious eye disease. Despite the severity of the disease caused by EV70, its structure is unknown. Here, we present the structures of the EV70 virion, altered particle, and empty capsid determined by cryo-electron microscopy. The capsid of EV70 is composed of the subunits VP1, VP2, VP3, and VP4. The partially collapsed hydrophobic pocket located in VP1 of the EV70 virion is not occupied by a pocket factor, which is commonly present in other enteroviruses. Nevertheless, we show that the pocket can be targeted by the antiviral compounds WIN51711 and pleconaril, which block virus infection. The inhibitors prevent genome release by stabilizing EV70 particles. Knowledge of the structures of complexes of EV70 with inhibitors will enable the development of capsid-binding therapeutics against this virus. IMPORTANCE Globally distributed enterovirus 70 (EV70) causes local outbreaks of acute hemorrhagic conjunctivitis. The discharge from infected eyes enables the high-efficiency transmission of EV70 in overcrowded areas with low hygienic standards. Currently, only symptomatic treatments are available. We determined the structures of EV70 in its native form, the genome release intermediate, and the empty capsid resulting from genome release. Furthermore, we elucidated the structures of EV70 in complex with two inhibitors that block virus infection, and we describe the mechanism of their binding to the virus capsid. These results enable the development of therapeutics against EV70.


Asunto(s)
Antivirales , Cápside , Enterovirus Humano D , Antivirales/farmacología , Cápside/ultraestructura , Proteínas de la Cápside , Conjuntivitis Hemorrágica Aguda/virología , Microscopía por Crioelectrón , Enterovirus Humano D/efectos de los fármacos , Enterovirus Humano D/ultraestructura , Humanos , Oxadiazoles/farmacología , Oxazoles/farmacología , Virión/efectos de los fármacos , Virión/ultraestructura
14.
J Gen Virol ; 103(5)2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35506996

RESUMEN

The family Potyviridae includes plant viruses with single-stranded, positive-sense RNA genomes of 8-11 kb and flexuous filamentous particles 650-950 nm long and 11-20 nm wide. Genera in the family are distinguished by the host range, genomic features and phylogeny of the member viruses. Most genomes are monopartite, but those of members of the genus Bymovirus are bipartite. Some members cause serious disease epidemics in cultivated plants. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the family Potyviridae, which is available at ictv.global/report/potyviridae.


Asunto(s)
Genoma Viral , Filogenia , Enfermedades de las Plantas/virología , Potyviridae/clasificación , Potyviridae/genética , Especificidad del Huésped , Virus de Plantas/clasificación , Virus de Plantas/genética , Plantas , ARN Viral/genética , Virión/genética , Virión/ultraestructura , Replicación Viral
15.
Viruses ; 14(4)2022 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-35458522

RESUMEN

Tick-borne encephalitis virus (TBEV) is a pathogenic, enveloped, positive-stranded RNA virus in the family Flaviviridae. Structural studies of flavivirus virions have primarily focused on mosquito-borne species, with only one cryo-electron microscopy (cryo-EM) structure of a tick-borne species published. Here, we present a 3.3 Å cryo-EM structure of the TBEV virion of the Kuutsalo-14 isolate, confirming the overall organisation of the virus. We observe conformational switching of the peripheral and transmembrane helices of M protein, which can explain the quasi-equivalent packing of the viral proteins and highlights their importance in stabilising membrane protein arrangement in the virion. The residues responsible for M protein interactions are highly conserved in TBEV but not in the structurally studied Hypr strain, nor in mosquito-borne flaviviruses. These interactions may compensate for the lower number of hydrogen bonds between E proteins in TBEV compared to the mosquito-borne flaviviruses. The structure reveals two lipids bound in the E protein which are important for virus assembly. The lipid pockets are comparable to those recently described in mosquito-borne Zika, Spondweni, Dengue, and Usutu viruses. Our results thus advance the understanding of tick-borne flavivirus architecture and virion-stabilising interactions.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas , Encefalitis Transmitida por Garrapatas , Animales , Microscopía por Crioelectrón , Culicidae , Virus de la Encefalitis Transmitidos por Garrapatas/genética , Virus de la Encefalitis Transmitidos por Garrapatas/ultraestructura , Proteínas Virales/metabolismo , Virión/metabolismo , Virión/ultraestructura , Virus Zika/metabolismo , Infección por el Virus Zika
16.
Cell ; 185(4): 641-653.e17, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-35123651

RESUMEN

HIV-1 Env mediates viral entry into host cells and is the sole target for neutralizing antibodies. However, Env structure and organization in its native virion context has eluded detailed characterization. Here, we used cryo-electron tomography to analyze Env in mature and immature HIV-1 particles. Immature particles showed distinct Env positioning relative to the underlying Gag lattice, providing insights into long-standing questions about Env incorporation. A 9.1-Å sub-tomogram-averaged reconstruction of virion-bound Env in conjunction with structural mass spectrometry revealed unexpected features, including a variable central core of the gp41 subunit, heterogeneous glycosylation between protomers, and a flexible stalk that allows Env tilting and variable exposure of neutralizing epitopes. Together, our results provide an integrative understanding of HIV assembly and structural variation in Env antigen presentation.


Asunto(s)
Microscopía por Crioelectrón , Tomografía con Microscopio Electrónico , Virión/ultraestructura , Productos del Gen env del Virus de la Inmunodeficiencia Humana/ultraestructura , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/ultraestructura , 2,2'-Dipiridil/análogos & derivados , 2,2'-Dipiridil/farmacología , Secuencia de Aminoácidos , Disulfuros/farmacología , Epítopos/química , Células HEK293 , Proteína gp41 de Envoltorio del VIH/química , Humanos , Espectrometría de Masas de Intercambio de Hidrógeno-Deuterio , Modelos Moleculares , Pruebas de Neutralización , Péptidos/química , Polisacáridos/química , Dominios Proteicos , Estructura Secundaria de Proteína , Subunidades de Proteína/química , Productos del Gen env del Virus de la Inmunodeficiencia Humana/química
17.
Nat Commun ; 13(1): 868, 2022 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-35165285

RESUMEN

SARS-CoV-2 infection is a major global public health concern with incompletely understood pathogenesis. The SARS-CoV-2 spike (S) glycoprotein comprises a highly conserved free fatty acid binding pocket (FABP) with unknown function and evolutionary selection advantage1,2. Deciphering FABP impact on COVID-19 progression is challenged by the heterogenous nature and large molecular variability of live virus. Here we create synthetic minimal virions (MiniVs) of wild-type and mutant SARS-CoV-2 with precise molecular composition and programmable complexity by bottom-up assembly. MiniV-based systematic assessment of S free fatty acid (FFA) binding reveals that FABP functions as an allosteric regulatory site enabling adaptation of SARS-CoV-2 immunogenicity to inflammation states via binding of pro-inflammatory FFAs. This is achieved by regulation of the S open-to-close equilibrium and the exposure of both, the receptor binding domain (RBD) and the SARS-CoV-2 RGD motif that is responsible for integrin co-receptor engagement. We find that the FDA-approved drugs vitamin K and dexamethasone modulate S-based cell binding in an FABP-like manner. In inflammatory FFA environments, neutralizing immunoglobulins from human convalescent COVID-19 donors lose neutralization activity. Empowered by our MiniV technology, we suggest a conserved mechanism by which SARS-CoV-2 dynamically couples its immunogenicity to the host immune response.


Asunto(s)
COVID-19/inmunología , Ácidos Grasos/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Virión/inmunología , Células A549 , Sitio Alostérico/genética , Secuencia de Aminoácidos , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Sitios de Unión/genética , COVID-19/metabolismo , COVID-19/virología , Células Cultivadas , Microscopía por Crioelectrón/métodos , Tomografía con Microscopio Electrónico/métodos , Proteínas de Unión a Ácidos Grasos/inmunología , Proteínas de Unión a Ácidos Grasos/metabolismo , Ácidos Grasos/metabolismo , Humanos , Células MCF-7 , Microscopía Confocal/métodos , Unión Proteica , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiología , Homología de Secuencia de Aminoácido , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Virión/metabolismo , Virión/ultraestructura
18.
Viruses ; 14(2)2022 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-35215818

RESUMEN

The coat proteins (CPs) of single-stranded RNA bacteriophages (ssRNA phages) directly assemble around the genomic RNA (gRNA) to form a near-icosahedral capsid with a single maturation protein (Mat) that binds the gRNA and interacts with the retractile pilus during infection of the host. Understanding the assembly of ssRNA phages is essential for their use in biotechnology, such as RNA protection and delivery. Here, we present the complete gRNA model of the ssRNA phage Qß, revealing that the 3' untranslated region binds to the Mat and the 4127 nucleotides fold domain-by-domain, and is connected through long-range RNA-RNA interactions, such as kissing loops. Thirty-three operator-like RNA stem-loops are located and primarily interact with the asymmetric A/B CP-dimers, suggesting a pathway for the assembly of the virions. Additionally, we have discovered various forms of the virus-like particles (VLPs), including the canonical T = 3 icosahedral, larger T = 4 icosahedral, prolate, oblate forms, and a small prolate form elongated along the 3-fold axis. These particles are all produced during a normal infection, as well as when overexpressing the CPs. When overexpressing the shorter RNA fragments encoding only the CPs, we observed an increased percentage of the smaller VLPs, which may be sufficient to encapsidate a shorter RNA.


Asunto(s)
Bacteriófagos/fisiología , Virión/fisiología , Ensamble de Virus , Bacteriófagos/química , Bacteriófagos/genética , Bacteriófagos/ultraestructura , Cápside/metabolismo , Cápside/ultraestructura , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Microscopía por Crioelectrón , Modelos Moleculares , ARN Viral/química , ARN Viral/genética , ARN Viral/metabolismo , Virión/química , Virión/genética , Virión/ultraestructura
19.
Viruses ; 14(2)2022 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-35215918

RESUMEN

Getah virus (GETV) is a member of the alphavirus genus, and it infects a variety of animal species, including horses, pigs, cattle, and foxes. Human infection with this virus has also been reported. The structure of GETV has not yet been determined. In this study, we report the cryo-EM structure of GETV at a resolution of 3.5 Å. This structure reveals conformational polymorphism of the envelope glycoproteins E1 and E2 at icosahedral 3-fold and quasi-3-fold axes, which is believed to be a necessary organization in forming a curvature surface of virions. In our density map, three extra densities are identified, one of which is believed a "pocket factor"; the other two are located by domain D of E2, and they may maintain the stability of E1/E2 heterodimers. We also identify three N-glycosylations at E1 N141, E2 N200, and E2 N262, which might be associated with receptor binding and membrane fusion. The resolving of the structure of GETV provides new insights into the structure and assembly of alphaviruses and lays a basis for studying the differences of biology and pathogenicity between arthritogenic and encephalitic alphaviruses.


Asunto(s)
Infecciones por Alphavirus/veterinaria , Infecciones por Alphavirus/virología , Alphavirus/fisiología , Alphavirus/ultraestructura , Ensamble de Virus , Alphavirus/clasificación , Alphavirus/genética , Animales , Bovinos/virología , Microscopía por Crioelectrón , Dimerización , Zorros/virología , Caballos/virología , Humanos , Modelos Moleculares , Filogenia , Porcinos/virología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Virión/clasificación , Virión/genética , Virión/fisiología , Virión/ultraestructura
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA