Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.964
Filtrar
1.
J Virol ; 98(3): e0153623, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38315014

RESUMO

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.


Assuntos
Vírus da Febre Suína Africana , Suínos , Proteínas Estruturais Virais , Animais , Febre Suína Africana/virologia , Vírus da Febre Suína Africana/química , Vírus da Febre Suína Africana/crescimento & desenvolvimento , Vírus da Febre Suína Africana/patogenicidade , Vírus da Febre Suína Africana/ultraestrutura , Microscopia Crioeletrônica , Relação Estrutura-Atividade , Suínos/virologia , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/metabolismo , Proteínas Estruturais Virais/ultraestrutura , Vírion/química , Vírion/metabolismo , Vírion/ultraestrutura , Virulência
2.
Nature ; 623(7989): 1026-1033, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37993716

RESUMO

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.


Assuntos
Antígenos CD4 , Membrana Celular , Proteína gp120 do Envelope de HIV , HIV-1 , Multimerização Proteica , Humanos , Vacinas contra a AIDS/química , Vacinas contra a AIDS/imunologia , Capsídeo/química , Capsídeo/metabolismo , Capsídeo/ultraestrutura , Antígenos CD4/química , Antígenos CD4/metabolismo , Antígenos CD4/ultraestrutura , Membrana Celular/química , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Microscopia Crioeletrônica , Tomografia com Microscopia Eletrônica , Anticorpos Anti-HIV/imunologia , Proteína gp120 do Envelope de HIV/química , Proteína gp120 do Envelope de HIV/metabolismo , Proteína gp120 do Envelope de HIV/ultraestrutura , Infecções por HIV/virologia , HIV-1/química , HIV-1/ultraestrutura , Vírion/química , Vírion/metabolismo , Vírion/ultraestrutura
3.
Nature ; 617(7960): 409-416, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37138077

RESUMO

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.


Assuntos
Vírus de DNA , Intestinos , Proteínas Virais , Vírion , Humanos , Capsídeo/química , Capsídeo/metabolismo , Capsídeo/ultraestrutura , Microscopia Crioeletrônica , Vírus de DNA/química , Vírus de DNA/classificação , Vírus de DNA/isolamento & purificação , Vírus de DNA/metabolismo , Vírus de DNA/ultraestrutura , Vírion/química , Vírion/metabolismo , Vírion/ultraestrutura , Montagem de Vírus , Intestinos/microbiologia , Intestinos/virologia , Proteínas Virais/química , Proteínas Virais/metabolismo , Proteínas Virais/ultraestrutura , Desdobramento de Proteína , Dobramento de Proteína
4.
Phys Chem Chem Phys ; 25(18): 12882-12890, 2023 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-37165855

RESUMO

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.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/metabolismo , Microscopia Crioeletrônica , Glicoproteína da Espícula de Coronavírus/metabolismo , Simulação de Dinâmica Molecular , Vírion/metabolismo , Vírion/ultraestrutura
5.
Viruses ; 14(12)2022 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-36560790

RESUMO

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.


Assuntos
COVID-19 , SARS-CoV-2 , Animais , Chlorocebus aethiops , Humanos , Células Vero , Pandemias , Vírion/ultraestrutura
6.
J Virol ; 96(24): e0136722, 2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36448797

RESUMO

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.


Assuntos
Cátions , Enterovirus Humano B , Humanos , Albuminas/farmacologia , Proteínas do Capsídeo/metabolismo , Cátions/farmacologia , Microscopia Crioeletrônica , Endossomos/metabolismo , Enterovirus Humano B/efeitos dos fármacos , Enterovirus Humano B/genética , Enterovirus Humano B/ultraestrutura , Infecções por Enterovirus/patologia , Infecções por Enterovirus/virologia , RNA/metabolismo , Vírion/efeitos dos fármacos , Vírion/metabolismo , Vírion/ultraestrutura , Genoma Viral
7.
J Virol ; 96(16): e0062722, 2022 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-35924923

RESUMO

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.


Assuntos
Anticorpos Amplamente Neutralizantes , Proteínas do Capsídeo , Epitopos de Linfócito B , Rotavirus , Vacinas Atenuadas , Vírion , Animais , Anticorpos Amplamente Neutralizantes/imunologia , Anticorpos Amplamente Neutralizantes/ultraestrutura , Proteínas do Capsídeo/química , Proteínas do Capsídeo/imunologia , Proteínas do Capsídeo/ultraestrutura , Microscopia Crioeletrônica , Epitopos de Linfócito B/imunologia , Epitopos de Linfócito B/ultraestrutura , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Fragmentos Fab das Imunoglobulinas/ultraestrutura , Camundongos , Conformação Proteica , Ratos , Rotavirus/química , Rotavirus/classificação , Rotavirus/imunologia , Rotavirus/fisiologia , Inoculações Seriadas , Vacinas Atenuadas/química , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/metabolismo , Vírion/imunologia , Vírion/metabolismo , Vírion/ultraestrutura
8.
J Virol ; 96(17): e0060422, 2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-35939401

RESUMO

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.


Assuntos
Antivirais , Capsídeo , Enterovirus Humano D , Antivirais/farmacologia , Capsídeo/ultraestrutura , Proteínas do Capsídeo , Conjuntivite Hemorrágica Aguda/virologia , Microscopia Crioeletrônica , Enterovirus Humano D/efeitos dos fármacos , Enterovirus Humano D/ultraestrutura , Humanos , Oxidiazóis/farmacologia , Oxazóis/farmacologia , Vírion/efeitos dos fármacos , Vírion/ultraestrutura
9.
J Gen Virol ; 103(5)2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35506996

RESUMO

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.


Assuntos
Genoma Viral , Filogenia , Doenças das Plantas/virologia , Potyviridae/classificação , Potyviridae/genética , Especificidade de Hospedeiro , Vírus de Plantas/classificação , Vírus de Plantas/genética , Plantas , RNA Viral/genética , Vírion/genética , Vírion/ultraestrutura , Replicação Viral
10.
Viruses ; 14(4)2022 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-35458522

RESUMO

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.


Assuntos
Vírus da Encefalite Transmitidos por Carrapatos , Encefalite Transmitida por Carrapatos , Animais , Microscopia Crioeletrônica , Culicidae , Vírus da Encefalite Transmitidos por Carrapatos/genética , Vírus da Encefalite Transmitidos por Carrapatos/ultraestrutura , Proteínas Virais/metabolismo , Vírion/metabolismo , Vírion/ultraestrutura , Zika virus/metabolismo , Infecção por Zika virus
11.
Cell ; 185(4): 641-653.e17, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-35123651

RESUMO

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.


Assuntos
Microscopia Crioeletrônica , Tomografia com Microscopia Eletrônica , Vírion/ultraestrutura , Produtos do Gene env do Vírus da Imunodeficiência Humana/ultraestrutura , Produtos do Gene gag do Vírus da Imunodeficiência Humana/ultraestrutura , 2,2'-Dipiridil/análogos & derivados , 2,2'-Dipiridil/farmacologia , Sequência de Aminoácidos , Dissulfetos/farmacologia , Epitopos/química , Células HEK293 , Proteína gp41 do Envelope de HIV/química , Humanos , Espectrometria de Massa com Troca Hidrogênio-Deutério , Modelos Moleculares , Testes de Neutralização , Peptídeos/química , Polissacarídeos/química , Domínios Proteicos , Estrutura Secundária de Proteína , Subunidades Proteicas/química , Produtos do Gene env do Vírus da Imunodeficiência Humana/química
12.
Nat Commun ; 13(1): 868, 2022 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-35165285

RESUMO

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.


Assuntos
COVID-19/imunologia , Ácidos Graxos/imunologia , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Vírion/imunologia , Células A549 , Sítio Alostérico/genética , Sequência de Aminoácidos , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Sítios de Ligação/genética , COVID-19/metabolismo , COVID-19/virologia , Células Cultivadas , Microscopia Crioeletrônica/métodos , Tomografia com Microscopia Eletrônica/métodos , Proteínas de Ligação a Ácido Graxo/imunologia , Proteínas de Ligação a Ácido Graxo/metabolismo , Ácidos Graxos/metabolismo , Humanos , Células MCF-7 , Microscopia Confocal/métodos , Ligação Proteica , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiologia , Homologia de Sequência de Aminoácidos , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/metabolismo , Vírion/metabolismo , Vírion/ultraestrutura
13.
Viruses ; 14(2)2022 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-35215818

RESUMO

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.


Assuntos
Bacteriófagos/fisiologia , Vírion/fisiologia , Montagem de Vírus , Bacteriófagos/química , Bacteriófagos/genética , Bacteriófagos/ultraestrutura , Capsídeo/metabolismo , Capsídeo/ultraestrutura , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Microscopia Crioeletrônica , Modelos Moleculares , RNA Viral/química , RNA Viral/genética , RNA Viral/metabolismo , Vírion/química , Vírion/genética , Vírion/ultraestrutura
14.
Viruses ; 14(2)2022 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-35215918

RESUMO

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.


Assuntos
Infecções por Alphavirus/veterinária , Infecções por Alphavirus/virologia , Alphavirus/fisiologia , Alphavirus/ultraestrutura , Montagem de Vírus , Alphavirus/classificação , Alphavirus/genética , Animais , Bovinos/virologia , Microscopia Crioeletrônica , Dimerização , Raposas/virologia , Cavalos/virologia , Humanos , Modelos Moleculares , Filogenia , Suínos/virologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Vírion/classificação , Vírion/genética , Vírion/fisiologia , Vírion/ultraestrutura
15.
Cell ; 185(2): 250-265.e16, 2022 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-35021064

RESUMO

Methods to deliver gene editing agents in vivo as ribonucleoproteins could offer safety advantages over nucleic acid delivery approaches. We report the development and application of engineered DNA-free virus-like particles (eVLPs) that efficiently package and deliver base editor or Cas9 ribonucleoproteins. By engineering VLPs to overcome cargo packaging, release, and localization bottlenecks, we developed fourth-generation eVLPs that mediate efficient base editing in several primary mouse and human cell types. Using different glycoproteins in eVLPs alters their cellular tropism. Single injections of eVLPs into mice support therapeutic levels of base editing in multiple tissues, reducing serum Pcsk9 levels 78% following 63% liver editing, and partially restoring visual function in a mouse model of genetic blindness. In vitro and in vivo off-target editing from eVLPs was virtually undetected, an improvement over AAV or plasmid delivery. These results establish eVLPs as promising vehicles for therapeutic macromolecule delivery that combine key advantages of both viral and nonviral delivery.


Assuntos
Sistemas de Liberação de Medicamentos , Engenharia Genética , Proteínas/uso terapêutico , Vírion/genética , Animais , Sequência de Bases , Cegueira/genética , Cegueira/terapia , Encéfalo/metabolismo , DNA/metabolismo , Modelos Animais de Doenças , Fibroblastos/metabolismo , Edição de Genes , Células HEK293 , Humanos , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Pró-Proteína Convertase 9/metabolismo , Epitélio Pigmentado da Retina/patologia , Retroviridae , Vírion/ultraestrutura , Visão Ocular
16.
Virology ; 568: 23-30, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35077959

RESUMO

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.


Assuntos
Eletroforese Capilar , Interações Hidrofóbicas e Hidrofílicas , Vírion/química , Algoritmos , Sequência de Aminoácidos , Bacteriófagos/química , Humanos , Modelos Teóricos , Dodecilsulfato de Sódio , Proteínas Virais/química , Vírion/isolamento & purificação , Vírion/ultraestrutura
17.
J Virol ; 96(4): e0163021, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-34908445

RESUMO

The matrix protein (M1) of influenza A virus plays an important role in replication, assembly, and budding. A previous study found that aspartic acid (D) at position 30 and alanine (A) at position 215 of M1 contribute to the high pathogenicity of H5N1 viruses in mice, and double mutations of D to asparagine (N) at position 30 (D30N) and A to threonine (T) at position 215 (A215T) in M1 dramatically attenuate H5N1 viruses in mice. However, the underlying mechanisms by which these M1 mutations attenuate the virulence of H5N1 viruses are unknown. Here, we found that the amino acid mutation A215T eliminates the SUMOylation of M1 by reducing its interaction with the host SUMO1 protein, significantly reducing the stability of M1, slowing the export of the M1-vRNP complex from the nucleus to the cytoplasm, and reducing viral replication in MDCK cells. We further found that the D30N mutation in M1 alters the shape of progeny viruses from filamentous to spherical virions. Our findings reveal an essential role for M1 215A SUMOylation and M1 30D-related filamentous morphology in the pathogenesis of avian influenza viruses, which could be targeted in novel antiviral drug designs. IMPORTANCE Identification of the pathogenic mechanism of highly pathogenic avian influenza viruses in mammals is helpful to develop novel anti-influenza virus strategies. Two amino acid mutations (D30N and A215T) in M1 were found to collectively attenuate H5N1 influenza viruses in mice, but the underlying mechanism remained unknown. This study found that the A215T mutation significantly decreases the SUMOylation of M1, which in turn attenuates the replication of H5N1 virus in mammalian cells. The D30N mutation in M1 was found to change the virion shape from filamentous to spherical. These findings are important for understanding the molecular mechanism of virulence of highly pathogenic avian influenza viruses in mammals.


Assuntos
Virus da Influenza A Subtipo H5N1/fisiologia , Virus da Influenza A Subtipo H5N1/patogenicidade , Infecções por Orthomyxoviridae/virologia , Proteínas da Matriz Viral/metabolismo , Replicação Viral , Transporte Ativo do Núcleo Celular , Animais , Núcleo Celular/metabolismo , Cães , Virus da Influenza A Subtipo H5N1/metabolismo , Virus da Influenza A Subtipo H5N1/ultraestrutura , Células Madin Darby de Rim Canino , Camundongos , Mutação , Infecções por Orthomyxoviridae/metabolismo , Estabilidade Proteica , Ribonucleoproteínas/metabolismo , Sumoilação , Proteínas da Matriz Viral/genética , Vírion/ultraestrutura , Virulência/genética , Replicação Viral/genética
18.
J Cell Mol Med ; 26(1): 25-34, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34904376

RESUMO

Transmission electron microscopy has historically been indispensable for virology research, as it offers unique insight into virus function. In the past decade, as cryo-electron microscopy (cryo-EM) has matured and become more accessible, we have been able to peer into the structure of viruses at the atomic level and understand how they interact with the host cell, with drugs or with antibodies. Perhaps, there was no time in recent history where cryo-EM was more needed, as SARS-CoV-2 has spread around the globe, causing millions of deaths and almost unquantifiable economic devastation. In this concise review, we aim to mark the most important contributions of cryo-EM to understanding the structure and function of SARS-CoV-2 proteins, from surface spikes to the virus core and from virus-receptor interactions to antibody binding.


Assuntos
Enzima de Conversão de Angiotensina 2/química , Anticorpos Antivirais/química , Vacinas contra COVID-19/química , COVID-19/prevenção & controle , Receptores Virais/química , Glicoproteína da Espícula de Coronavírus/química , Enzima de Conversão de Angiotensina 2/imunologia , Enzima de Conversão de Angiotensina 2/metabolismo , Anticorpos Antivirais/biossíntese , COVID-19/imunologia , COVID-19/virologia , Vacinas contra COVID-19/administração & dosagem , Vacinas contra COVID-19/biossíntese , Microscopia Crioeletrônica , Epitopos/química , Epitopos/imunologia , Epitopos/metabolismo , Humanos , Modelos Moleculares , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Receptores Virais/imunologia , Receptores Virais/metabolismo , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/patogenicidade , SARS-CoV-2/ultraestrutura , Serina Endopeptidases/química , Serina Endopeptidases/imunologia , Serina Endopeptidases/metabolismo , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Vírion/efeitos dos fármacos , Vírion/patogenicidade , Vírion/ultraestrutura
19.
Med Mol Morphol ; 55(1): 60-67, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34825978

RESUMO

SARS-CoV-2 is the cause of COVID-19. The three-dimensional morphology of viral particles existing and multiplying in infected cells has not been established by electron tomography, which is different from cryo-electron tomography using frozen samples. In this study, we establish the morphological structure of SARS-CoV-2 particles by three-dimensional reconstruction of images obtained by electron tomography and transmission electron microscopy of biological samples embedded in epoxy resin. The characteristic roots of spike structures were found to be arranged at the surface of a virion covered with an envelope. A high-electron-density structure that appears to be a nucleocapsid was observed inside the envelope of the virion on three-dimensional images reconstructed by electron tomography. The SARS-CoV-2 particles that budded in the vacuoles in the cytoplasm were morphologically identical to those found outside the cells, suggesting that mature and infectious SARS-CoV-2 particles were already produced in the vacuoles. Here, we show the three-dimensional morphological structure of SARS-CoV-2 particles reconstructed by electron tomography. To control infection, inhibition of viral release from vacuoles would be a new target in the development of prophylactic agents against SARS-CoV-2.


Assuntos
Tomografia com Microscopia Eletrônica , SARS-CoV-2 , COVID-19 , Humanos , Imageamento Tridimensional , SARS-CoV-2/ultraestrutura , Vírion/ultraestrutura
20.
J Virol ; 96(3): e0166721, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-34787458

RESUMO

African swine fever virus (ASFV) is a complex nucleocytoplasmic large DNA virus that causes African swine fever, a lethal hemorrhagic disease that currently threatens the pig industry. Recent studies have identified the viral structural proteins of infectious ASFV particles. However, the functional roles of several ASFV structural proteins remain largely unknown. Here, we characterized the function of the ASFV structural protein H240R (pH240R) in virus morphogenesis. pH240R was identified as a capsid protein by using immunoelectron microscopy and interacted with the major capsid protein p72 by pulldown assays. Using a recombinant ASFV, ASFV-ΔH240R, with the H240R gene deleted from the wild-type ASFV (ASFV-WT) genome, we revealed that the infectious progeny virus titers were reduced by approximately 2.0 logs compared with those of ASFV-WT. Furthermore, we demonstrated that the growth defect was due to the generation of noninfectious particles with a higher particle-to-infectious titer ratio in ASFV-ΔH240R-infected primary porcine alveolar macrophages (PAMs) than in those infected with ASFV-WT. Importantly, we found that pH240R did not affect virus-cell binding, endocytosis, or egress but did affect ASFV assembly; noninfectious virions containing large aberrant tubular and bilobulate structures comprised nearly 98% of all virions observed in ASFV-ΔH240R-infected PAMs by electron microscopy. Notably, we demonstrated that ASFV-ΔH240R infection induced high-level expression of inflammatory cytokines in PAMs. Collectively, we show for the first time that pH240R is essential for ASFV icosahedral capsid formation and infectious particle production. Also, these results highlight the importance of pH240R in ASFV morphogenesis and provide a novel target for the development of ASF vaccines and antivirals. IMPORTANCE African swine fever is a lethal hemorrhagic disease of global concern that is caused by African swine fever virus (ASFV). Despite extensive research, there exist relevant gaps in knowledge of the fundamental biology of the viral life cycle. In this study, we identified pH240R as a capsid protein that interacts with the major capsid protein p72. Furthermore, we showed that pH240R was required for the efficient production of infectious progeny virions as indicated by the H240R-deleted ASFV mutant (ASFV-ΔH240R). More specifically, pH240R directs the morphogenesis of ASFV toward the icosahedral capsid in the process of assembly. In addition, ASFV-ΔH240R infection induced high-level expression of inflammatory cytokines in primary porcine alveolar macrophages. Our results elucidate the role of pH240R in the process of ASFV assembly, which may instruct future research on effective vaccines or antiviral strategies.


Assuntos
Vírus da Febre Suína Africana/fisiologia , Febre Suína Africana/genética , Febre Suína Africana/metabolismo , Proteínas do Capsídeo/genética , Citocinas/metabolismo , Macrófagos/metabolismo , Deleção de Sequência , Febre Suína Africana/patologia , Vírus da Febre Suína Africana/ultraestrutura , Sequência de Aminoácidos , Animais , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Citocinas/genética , Suscetibilidade a Doenças/imunologia , Perfilação da Expressão Gênica , Regulação Viral da Expressão Gênica , Genoma Viral , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Macrófagos/imunologia , Suínos , Vírion/ultraestrutura , Internalização do Vírus , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...