Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
1.
J Virol ; 98(3): e0157623, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38323814

RESUMO

Adenovirus (AdV) infection of the respiratory epithelium is common but poorly understood. Human AdV species C types, such as HAdV-C5, utilize the Coxsackie-adenovirus receptor (CAR) for attachment and subsequently integrins for entry. CAR and integrins are however located deep within the tight junctions in the mucosa where they would not be easily accessible. Recently, a model for CAR-independent AdV entry was proposed. In this model, human lactoferrin (hLF), an innate immune protein, aids the viral uptake into epithelial cells by mediating interactions between the major capsid protein, hexon, and yet unknown host cellular receptor(s). However, a detailed understanding of the molecular interactions driving this mechanism is lacking. Here, we present a new cryo-EM structure of HAdV-5C hexon at high resolution alongside a hybrid structure of HAdV-5C hexon complexed with human lactoferrin (hLF). These structures reveal the molecular determinants of the interaction between hLF and HAdV-C5 hexon. hLF engages hexon primarily via its N-terminal lactoferricin (Lfcin) region, interacting with hexon's hypervariable region 1 (HVR-1). Mutational analyses pinpoint critical Lfcin contacts and also identify additional regions within hLF that critically contribute to hexon binding. Our study sheds more light on the intricate mechanism by which HAdV-C5 utilizes soluble hLF/Lfcin for cellular entry. These findings hold promise for advancing gene therapy applications and inform vaccine development. IMPORTANCE: Our study delves into the structural aspects of adenovirus (AdV) infections, specifically HAdV-C5 in the respiratory epithelium. It uncovers the molecular details of a novel pathway where human lactoferrin (hLF) interacts with the major capsid protein, hexon, facilitating viral entry, and bypassing traditional receptors such as CAR and integrins. The study's cryo-EM structures reveal how hLF engages hexon, primarily through its N-terminal lactoferricin (Lfcin) region and hexon's hypervariable region 1 (HVR-1). Mutational analyses identify critical Lfcin contacts and other regions within hLF vital for hexon binding. This structural insight sheds light on HAdV-C5's mechanism of utilizing soluble hLF/Lfcin for cellular entry, holding promise for gene therapy and vaccine development advancements in adenovirus research.


Assuntos
Adenovírus Humanos , Proteínas do Capsídeo , Lactoferrina , Receptores Virais , Internalização do Vírus , Humanos , Infecções por Adenovirus Humanos/metabolismo , Infecções por Adenovirus Humanos/virologia , Adenovírus Humanos/química , Adenovírus Humanos/genética , Adenovírus Humanos/metabolismo , Adenovírus Humanos/ultraestrutura , Sítios de Ligação/genética , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Proteínas do Capsídeo/ultraestrutura , Microscopia Crioeletrônica , Lactoferrina/química , Lactoferrina/genética , Lactoferrina/metabolismo , Lactoferrina/ultraestrutura , Modelos Biológicos , Mutação , Ligação Proteica , Receptores Virais/química , Receptores Virais/genética , Receptores Virais/metabolismo , Receptores Virais/ultraestrutura , Solubilidade , Mucosa Respiratória/citologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/virologia
2.
Virology ; 565: 22-28, 2022 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-34638006

RESUMO

Adeno-associated virus (AAV) are classified as non-enveloped ssDNA viruses. However, AAV capsids embedded within exosomes have been observed, and it has been suggested that the AAV membrane associated accessory protein (MAAP) may play a role in envelope-associated AAV (EA-AAV) capsid formation. Here, we observed and selected sufficient homogeneous EA-AAV capsids of AAV2, produced using the Sf9 baculoviral expression system, to determine the cryo-electron microscopy (cryo-EM) structure at 3.14 Å resolution. The reconstructed map confirmed that the EA-AAV capsid, showed no significant structural variation compared to the non-envelope capsid. In addition, the Sf9 expression system used implies the notion that MAAP may enhance exosome AAV encapsulation. Furthermore, we speculate that these EA-AAV capsids may have therapeutic benefits over the currently used non-envelope AAV capsids, with advantages in immune evasion and/or improved infectivity.


Assuntos
Proteínas do Capsídeo/ultraestrutura , Capsídeo/ultraestrutura , Dependovirus/ultraestrutura , Animais , Capsídeo/química , Proteínas do Capsídeo/química , Microscopia Crioeletrônica , Dependovirus/química , Exossomos , Evasão da Resposta Imune , Conformação Proteica , Células Sf9
3.
Nat Commun ; 12(1): 3226, 2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-34050170

RESUMO

Inositol hexakisphosphate (IP6) is an assembly cofactor for HIV-1. We report here that IP6 is also used for assembly of Rous sarcoma virus (RSV), a retrovirus from a different genus. IP6 is ~100-fold more potent at promoting RSV mature capsid protein (CA) assembly than observed for HIV-1 and removal of IP6 in cells reduces infectivity by 100-fold. Here, visualized by cryo-electron tomography and subtomogram averaging, mature capsid-like particles show an IP6-like density in the CA hexamer, coordinated by rings of six lysines and six arginines. Phosphate and IP6 have opposing effects on CA in vitro assembly, inducing formation of T = 1 icosahedrons and tubes, respectively, implying that phosphate promotes pentamer and IP6 hexamer formation. Subtomogram averaging and classification optimized for analysis of pleomorphic retrovirus particles reveal that the heterogeneity of mature RSV CA polyhedrons results from an unexpected, intrinsic CA hexamer flexibility. In contrast, the CA pentamer forms rigid units organizing the local architecture. These different features of hexamers and pentamers determine the structural mechanism to form CA polyhedrons of variable shape in mature RSV particles.


Assuntos
Proteínas do Capsídeo/metabolismo , Capsídeo/ultraestrutura , Ácido Fítico/metabolismo , Vírus do Sarcoma de Rous/ultraestrutura , Montagem de Vírus , Capsídeo/metabolismo , Proteínas do Capsídeo/isolamento & purificação , Proteínas do Capsídeo/ultraestrutura , Microscopia Crioeletrônica , Tomografia com Microscopia Eletrônica , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Modelos Moleculares , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Multimerização Proteica , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Vírus do Sarcoma de Rous/patogenicidade , Vírus do Sarcoma de Rous/fisiologia , Imagem Individual de Molécula , Transfecção , Liberação de Vírus
4.
Biochem Biophys Res Commun ; 559: 161-167, 2021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-33940388

RESUMO

VLPs are virus-like particles that comprise viral capsid proteins that can self-assemble and mimic the shape and size of real viral particles; however, because they do not contain genetic material they cannot infect host cells. VLPs have great potential as safe drug/vehicle candidates; therefore, they are gaining popularity in the field of preventive medicine and therapeutics. Indeed, extensive studies are underway to examine their role as carriers for immunization and as vehicles for delivery of therapeutic agents. Here, we examined the possibility of developing VLP-utilizing technology based on an efficient VLP production process and high-resolution structural analysis. Nicotiana benthamiana was used as an expression platform to produce the coat protein of the alfalfa mosaic virus (AMV-CP). About 250 mg/kg of rAMV-CP was produced from Nicotiana benthamiana leaves. Structural analysis revealed that the oligomeric status of rAMV-CP changed according to the composition and pH of the buffer. Size exclusion chromatography and electron microscopy analysis confirmed the optimal conditions for rAMV-CP VLP formation, and a 2.4 Å resolution structure was confirmed by cryo-EM analysis. Based on the efficient protein production, VLP manufacturing technology, and high-resolution structure presented herein, we suggest that rAMV-CP VLP is a useful platform for development of various new drugs.


Assuntos
Vírus do Mosaico da Alfafa/ultraestrutura , Proteínas do Capsídeo/ultraestrutura , Nicotiana/virologia , Vírus do Mosaico da Alfafa/química , Proteínas do Capsídeo/química , Microscopia Crioeletrônica , Modelos Moleculares , Conformação Proteica
5.
Cell Host Microbe ; 29(3): 448-462.e5, 2021 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-33539764

RESUMO

Enterovirus uncoating receptors bind at the surface depression ("canyon") that encircles each capsid vertex causing the release of a host-derived lipid called "pocket factor" that is buried in a hydrophobic pocket formed by the major viral capsid protein, VP1. Coxsackievirus and adenovirus receptor (CAR) is a universal uncoating receptor of group B coxsackieviruses (CVB). Here, we present five high-resolution cryoEM structures of CVB representing different stages of virus infection. Structural comparisons show that the CAR penetrates deeper into the canyon than other uncoating receptors, leading to a cascade of events: collapse of the VP1 hydrophobic pocket, high-efficiency release of the pocket factor and viral uncoating and genome release under neutral pH, as compared with low pH. Furthermore, we identified a potent therapeutic antibody that can neutralize viral infection by interfering with virion-CAR interactions, destabilizing the capsid and inducing virion disruption. Together, these results define the structural basis of CVB cell entry and antibody neutralization.


Assuntos
Microscopia Crioeletrônica , Enterovirus/metabolismo , Enterovirus/ultraestrutura , Animais , Anticorpos Neutralizantes , Capsídeo/metabolismo , Proteínas do Capsídeo/ultraestrutura , Enterovirus Humano B/metabolismo , Infecções por Enterovirus/imunologia , Infecções por Enterovirus/metabolismo , Infecções por Enterovirus/virologia , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Domínios e Motivos de Interação entre Proteínas , Receptores Virais , Vírion/metabolismo , Vírion/ultraestrutura , Desenvelopamento do Vírus
6.
Viruses ; 13(1)2021 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-33467558

RESUMO

Adenovirus (AdV) infection elicits a strong immune response with the production of neutralizing antibodies and opsonization by complement and coagulation factors. One anti-hexon neutralizing antibody, called 9C12, is known to activate the complement cascade, resulting in the deposition of complement component C4b on the capsid, and the neutralization of the virus. The mechanism of AdV neutralization by C4b is independent of downstream complement proteins and involves the blockage of the release of protein VI, which is required for viral escape from the endosome. To investigate the structural basis underlying how C4b blocks the uncoating of AdV, we built a model for the complex of human adenovirus type-5 (HAdV5) with 9C12, together with complement components C1 and C4b. This model positions C4b near the Arg-Gly-Asp (RGD) loops of the penton base. There are multiple amino acids in the RGD loop that might serve as covalent binding sites for the reactive thioester of C4b. Molecular dynamics simulations with a multimeric penton base and C4b indicated that stabilizing interactions may form between C4b and multiple RGD loops. We propose that C4b deposition on one RGD loop leads to the entanglement of C4b with additional RGD loops on the same penton base multimer and that this entanglement blocks AdV uncoating.


Assuntos
Adenoviridae/imunologia , Complemento C4/química , Complemento C4/imunologia , Modelos Moleculares , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/química , Anticorpos Antivirais/imunologia , Sítios de Ligação , Capsídeo/química , Capsídeo/metabolismo , Capsídeo/ultraestrutura , Proteínas do Capsídeo/química , Proteínas do Capsídeo/imunologia , Proteínas do Capsídeo/metabolismo , Proteínas do Capsídeo/ultraestrutura , Humanos , Imunoglobulina G/química , Imunoglobulina G/imunologia , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade
7.
Methods Mol Biol ; 2208: 149-165, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32856261

RESUMO

Nanoscale systems encapsulating biomacromolecules hold promise for cell and gene therapies. Common issues hampering progress include polydispersity, heterogeneity in size and shape, agglomeration, and poor stability. Much attention is given to the search of novel designs. However, reliable protocols for the validation of encapsulating systems in the continuum of their physicochemical properties, from design to ultrastructure, are lacking. Herein, we report electron microscopy protocols for biologically functional shell-like peptide capsids, which exhibit the physical characteristics of viruses including folding-mediated self-assembly, hollow shell morphology, and uniformity in size.


Assuntos
Proteínas do Capsídeo/ultraestrutura , Capsídeo/ultraestrutura , Microscopia Eletrônica/métodos , Peptídeos/química , Imageamento Tridimensional/métodos , Montagem de Vírus/fisiologia , Vírus/ultraestrutura
8.
Subcell Biochem ; 96: 503-518, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33252742

RESUMO

Human adenoviruses (HAdVs) are large (150 MDa), complex, nonenveloped dsDNA viruses that cause self-limiting respiratory, ocular and enteric infections. They are significant health hazard in young, elderly and immuno-compromised populations. Moreover, various adenoviruses (AdVs) of mammalian origin are being used as vectors in gene, vaccine and cancer therapies. Multiple copies of at least 13 different proteins, all in all ~2800 protein molecules, come together to form an adenovirus virion packaging the ~36 Kbp geome. The details of structural organization of the adenovirus capsid and underlying network of protein-protein interactions provide clues into designing the modified and novel adenovirus vectors with desired functionalities and/or targeting specificities. The advancements in 3D structure determination by cryo-electron microscopy (cryo-EM) in the past decade have enabled unveiling of the complex organization of adenovirus architecture at near atomic resolution. Specifically, these studies revealed the structures and the network of interactions involving cement/minor proteins in stabilizing the AdV icosahedral architecture, which appear to be mostly conserved among human adenoviruses. In this chapter, we describe the current state of knowledge on the structure and organization of human adenoviruses.


Assuntos
Adenovírus Humanos/química , Proteínas do Capsídeo/química , Capsídeo/química , Adenovírus Humanos/ultraestrutura , Capsídeo/ultraestrutura , Proteínas do Capsídeo/ultraestrutura , Microscopia Crioeletrônica , Humanos , Modelos Moleculares , Ligação Proteica
9.
PLoS Pathog ; 16(6): e1008588, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32584886

RESUMO

The human adenovirus type 5 (HAdV5) causes disease of the upper and lower respiratory tract. The early steps of HAdV5 entry up to genome replication in the host nucleus have been extensively studied. However, late stages of infection remain poorly understood. Here, we set out to elucidate the spatiotemporal orchestration of late adenovirus nuclear remodeling in living cells. We generated virus mutants expressing fluorescently tagged protein IX (pIX) and protein V (pV), a capsid and viral genome associated protein, respectively. We found that during progeny virion production both proteins localize to a membrane-less, nuclear compartment, which is highly impermeable such that in immunofluorescence microscopy antibodies can hardly penetrate it. We termed this compartment 'late virion accumulation compartment' (LVAC). Correlation between light- and electron microscopy revealed that the LVAC contains paracrystalline arrays of viral capsids that arrange tightly packed within a honeycomb-like organization of viral DNA. Live-cell microscopy as well as FRAP measurements showed that the LVAC is rigid and restricts diffusion of larger molecules, indicating that capsids are trapped inside.


Assuntos
Infecções por Adenovirus Humanos/metabolismo , Adenovírus Humanos/fisiologia , Proteínas do Capsídeo/metabolismo , DNA Viral/metabolismo , Vírion/metabolismo , Replicação Viral , Células A549 , Infecções por Adenovirus Humanos/genética , Infecções por Adenovirus Humanos/patologia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/ultraestrutura , DNA Viral/genética , Humanos , Vírion/genética
10.
J Struct Biol ; 211(2): 107547, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32522552

RESUMO

Adeno-associated viruses (AAVs) are widespread among vertebrates. AAVs isolated from bats display low capsid protein sequence identities (<60%) to AAV2, AAV5, and other primate AAVs. Here we report the first capsid structure of a non-primate AAV which was isolated from bats. The capsid structure of BtAAV-10HB (10HB) was determined by cryo-electron microscopy and three-dimensional image reconstruction to 3.03 Å resolution. Comparison of empty and genome-containing capsids showed that the capsid structures are almost identical except for an ordered nucleotide in a previously described nucleotide-binding pocket, the density in the 5-fold channel, and several amino acids with altered side chain conformations. Compared to other dependoparvoviruses, for example AAV2 and AAV5, 10HB displays unique structural features including insertions and deletions in capsid surface loops. Overall, the 10HB capsid structure superposes with an RMSD of 1.7 Å and 1.8 Å to AAV2 and AAV5, respectively. Currently all approved AAV human gene therapy biologics and vectors in clinical trials are based on primate isolates. However, pre-existing neutralizing antibodies in the human population represents a hurdle to their use. 10HB capsids are capable of packaging AAV2 vector genomes and thus have potential as gene delivery vectors. Significantly, a screen with human sera showed lack of recognition by the 10HB capsid. Thus, the different capsid surface of 10HB vectors likely renders it "invisible" to potential pre-existing neutralizing human anti-AAV antibodies especially because this virus or similar variants do not exist in primate populations.


Assuntos
Proteínas do Capsídeo/ultraestrutura , Capsídeo/ultraestrutura , Quirópteros/virologia , Dependovirus/ultraestrutura , Animais , Proteínas do Capsídeo/genética , Quirópteros/genética , Microscopia Crioeletrônica , Dependovirus/genética , Humanos , Modelos Moleculares , Ligação Proteica/genética
11.
J Virol ; 94(6)2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-31826994

RESUMO

Adeno-associated viruses (AAVs) from clade E are often used as vectors in gene delivery applications. This clade includes rhesus isolate 10 (AAVrh.10) and 39 (AAVrh.39) which, unlike representative AAV8, are capable of crossing the blood-brain barrier (BBB), thereby enabling the delivery of therapeutic genes to the central nervous system. Here, the capsid structures of AAV8, AAVrh.10 and AAVrh.39 have been determined by cryo-electron microscopy and three-dimensional image reconstruction to 3.08-, 2.75-, and 3.39-Šresolution, respectively, to enable a direct structural comparison. AAVrh.10 and AAVrh.39 are 98% identical in amino acid sequence but only ∼93.5% identical to AAV8. However, the capsid structures of all three viruses are similar, with only minor differences observed in the previously described surface variable regions, suggesting that specific residues S269 and N472, absent in AAV8, may confer the ability to cross the BBB in AAVrh.10 and AAVrh.39. Head-to-head comparison of empty and genome-containing particles showed DNA ordered in the previously described nucleotide-binding pocket, supporting the suggested role of this pocket in DNA packaging for the Dependoparvovirus The structural characterization of these viruses provides a platform for future vector engineering efforts toward improved gene delivery success with respect to specific tissue targeting, transduction efficiency, antigenicity, or receptor retargeting.IMPORTANCE Recombinant adeno-associated virus vectors (rAAVs), based on AAV8 and AAVrh.10, have been utilized in multiple clinical trials to treat different monogenetic diseases. The closely related AAVrh.39 has also shown promise in vivo As recently attained for other AAV biologics, e.g., Luxturna and Zolgensma, based on AAV2 and AAV9, respectively, the vectors in this study will likely gain U.S. Food and Drug Administration approval for commercialization in the near future. This study characterized the capsid structures of these clinical vectors at atomic resolution using cryo-electron microscopy and image reconstruction for comparative analysis. The analysis suggested two key residues, S269 and N472, as determinants of BBB crossing for AAVrh.10 and AAVrh.39, a feature utilized for central nervous system delivery of therapeutic genes. The structure information thus provides a platform for engineering to improve receptor retargeting or tissue specificity. These are important challenges in the field that need attention. Capsid structure information also provides knowledge potentially applicable for regulatory product approval.


Assuntos
Proteínas do Capsídeo/química , Proteínas do Capsídeo/ultraestrutura , Capsídeo/química , Capsídeo/ultraestrutura , Dependovirus/química , Sequência de Aminoácidos , Barreira Hematoencefálica , Proteínas do Capsídeo/genética , Microscopia Crioeletrônica , Dependovirus/genética , Terapia Genética , Vetores Genéticos , Células HEK293 , Humanos , Imageamento Tridimensional , Modelos Moleculares , Estados Unidos , United States Food and Drug Administration
12.
J Virol ; 94(2)2020 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-31694952

RESUMO

Members of the Tombusviridae family have highly similar structures, and yet there are important differences among them in host, transmission, and capsid stabilities. Viruses in the Tombusviridae family have single-stranded RNA (ssRNA) genomes with T=3 icosahedral protein shells with a maximum diameter of ∼340 Å. Each capsid protein is comprised of three domains: R (RNA binding), S (shell), and P (protruding). Between the R domain and S domain is the "arm" region that studies have shown to play a critical role in assembly. To better understand how the details of structural differences and similarities influence the Tombusviridae viral life cycles, the structures of cucumber leaf spot virus (CLSV; genus Aureusvirus) and red clover necrotic mosaic virus (RCNMV; genus Dianthovirus) were determined to resolutions of 3.2 Å and 2.9 Å, respectively, with cryo-electron microscopy and image reconstruction methods. While the shell domains had homologous structures, the stabilizing interactions at the icosahedral 3-fold axes and the R domains differed greatly. The heterogeneity in the R domains among the members of the Tombusviridae family is likely correlated with differences in the sizes and characteristics of the corresponding genomes. We propose that the changes in the R domain/RNA interactions evolved different arm domain interactions at the ß-annuli. For example, RCNMV has the largest genome and it appears to have created the necessary space in the capsid by evolving the shortest R domain. The resulting loss in RNA/R domain interactions may have been compensated for by increased intersubunit ß-strand interactions at the icosahedral 3-fold axes. Therefore, the R and arm domains may have coevolved to package different genomes within the conserved and rigid shell.IMPORTANCE Members of the Tombusviridae family have nearly identical shells, and yet they package genomes that range from 4.6 kb (monopartite) to 5.3 kb (bipartite) in size. To understand how this genome flexibility occurs within a rigidly conserved shell, we determined the high-resolution cryo-electron microscopy (cryo-EM) structures of cucumber leaf spot virus and red clover necrotic mosaic virus. In response to genomic size differences, it appears that the ssRNA binding (R) domain of the capsid diverged evolutionarily in order to recognize the different genomes. The next region, the "arm," seems to have also coevolved with the R domain to allow particle assembly via interactions at the icosahedral 3-fold axes. In addition, there are differences at the icosahedral 3-fold axes with regard to metal binding that are likely important for transmission and the viral life cycle.


Assuntos
Proteínas do Capsídeo/ultraestrutura , Capsídeo/ultraestrutura , Evolução Molecular , Tombusviridae/ultraestrutura , Microscopia Crioeletrônica , Nicotiana
13.
J Biol Chem ; 295(1): 1-12, 2020 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-31649031

RESUMO

African swine fever virus (ASFV) is a complex nucleocytoplasmic large DNA virus (NCLDV) that causes a devastating swine disease currently present in many countries of Africa, Europe, and Asia. Despite intense research efforts, relevant gaps in the architecture of the infectious virus particle remain. Here, we used single-particle cryo-EM to analyze the three-dimensional structure of the mature ASFV particle. Our results show that the ASFV virion, with a radial diameter of ∼2,080 Å, encloses a genome-containing nucleoid surrounded by two distinct icosahedral protein capsids and two lipoprotein membranes. The outer capsid forms a hexagonal lattice (triangulation number T = 277) composed of 8,280 copies of the double jelly-roll major capsid protein (MCP) p72, arranged in trimers displaying a pseudo-hexameric morphology, and of 60 copies of a penton protein at the vertices. The inner protein layer, organized as a T = 19 capsid, confines the core shell, and it is composed of the mature products derived from the ASFV polyproteins pp220 and pp62. Also, an icosahedral membrane lies between the two protein layers, whereas a pleomorphic envelope wraps the outer capsid. This high-level organization confers to ASFV a unique architecture among the NCLDVs that likely reflects the complexity of its infection process and may help explain current challenges in controlling it.


Assuntos
Vírus da Febre Suína Africana/ultraestrutura , Proteínas do Capsídeo/ultraestrutura , Capsídeo/ultraestrutura , Proteínas do Envelope Viral/ultraestrutura , Vírus da Febre Suína Africana/metabolismo , Animais , Proteínas do Capsídeo/química , Chlorocebus aethiops , Microscopia Crioeletrônica , Lipídeos/química , Multimerização Proteica , Células Vero , Proteínas do Envelope Viral/química
14.
Nat Commun ; 10(1): 5822, 2019 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-31862888

RESUMO

The HML2 (HERV-K) group constitutes the most recently acquired family of human endogenous retroviruses, with many proviruses less than one million years old. Many maintain intact open reading frames and provirus expression together with HML2 particle formation are observed in early stage human embryo development and are associated with pluripotency as well as inflammatory disease, cancers and HIV-1 infection. Here, we reconstruct the core structural protein (CA) of an HML2 retrovirus, assemble particles in vitro and employ single particle cryogenic electron microscopy (cryo-EM) to determine structures of four classes of CA Fullerene shell assemblies. These icosahedral and capsular assemblies reveal at high-resolution the molecular interactions that allow CA to form both pentamers and hexamers and show how invariant pentamers and structurally plastic hexamers associate to form the unique polyhedral structures found in retroviral cores.


Assuntos
Proteínas do Capsídeo/ultraestrutura , Capsídeo/ultraestrutura , Retrovirus Endógenos/ultraestrutura , Fulerenos/química , Estrutura Quaternária de Proteína , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/isolamento & purificação , Microscopia Crioeletrônica/métodos , Cristalografia por Raios X , Ressonância Magnética Nuclear Biomolecular , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/ultraestrutura , Imagem Individual de Molécula/métodos
15.
Nat Commun ; 10(1): 4840, 2019 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-31649265

RESUMO

Assembly of tailed bacteriophages and herpesviruses starts with formation of procapsids (virion precursors without DNA). Scaffolding proteins (SP) drive assembly by chaperoning the major capsid protein (MCP) to build an icosahedral lattice. Here we report near-atomic resolution cryo-EM structures of the bacteriophage SPP1 procapsid, the intermediate expanded procapsid with partially released SPs, and the mature capsid with DNA. In the intermediate state, SPs are bound only to MCP pentons and to adjacent subunits from hexons. SP departure results in the expanded state associated with unfolding of the MCP N-terminus and straightening of E-loops. The newly formed extensive inter-capsomere bonding appears to compensate for release of SPs that clasp MCP capsomeres together. Subsequent DNA packaging instigates bending of MCP A domain loops outwards, closing the hexons central opening and creating the capsid auxiliary protein binding interface. These findings provide a molecular basis for the sequential structural rearrangements during viral capsid maturation.


Assuntos
Bacteriófagos/ultraestrutura , Proteínas do Capsídeo/ultraestrutura , Capsídeo/ultraestrutura , Montagem de Vírus , Bacteriófagos/metabolismo , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Microscopia Crioeletrônica , Cristalografia por Raios X , Proteínas Estruturais Virais/metabolismo , Proteínas Estruturais Virais/ultraestrutura
16.
Hum Gene Ther ; 30(12): 1449-1460, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31530236

RESUMO

Adeno-associated viruses (AAVs) have been employed successfully as gene therapy vectors in treating various genetic diseases for almost two decades. However, transgene packaging is usually imperfect, and developing a rapid and accurate method for measuring the proportion of DNA encapsidation is an important step for improving the downstream process of large scale vector production. In this study, we used two-dimensional class averages and three-dimensional classes, intermediate outputs in the single particle cryo-electron microscopy (cryo-EM) image reconstruction pipeline, to determine the proportion of DNA-packaged and empty capsid populations. Two different preparations of AAV3 were analyzed to estimate the minimum number of particles required to be sampled by cryo-EM in order for robust calculation of the proportion of the full versus empty capsids in any given sample. Cost analysis applied to the minimum amount of data required for a valid ratio suggests that cryo-EM is an effective approach to analyze vector preparations.


Assuntos
Proteínas do Capsídeo/ultraestrutura , Capsídeo/ultraestrutura , Microscopia Crioeletrônica , Dependovirus/ultraestrutura , Proteínas do Capsídeo/genética , Dependovirus/genética , Vetores Genéticos/genética , Vetores Genéticos/uso terapêutico , Humanos , Vírion/genética , Vírion/ultraestrutura
17.
Nat Commun ; 10(1): 3891, 2019 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-31467275

RESUMO

Herpesviridae is a vast family of enveloped DNA viruses that includes eight distinct human pathogens, responsible for diseases that range from almost asymptomatic to severe and life-threatening. Epstein-Barr virus infects B-cells and epithelial cells, causing infectious mononucleosis, as well as a number of cancers. Epstein-Barr infection cannot be cured since neither vaccine nor antiviral drug treatments are available. All herpesviruses contain a linear double-stranded DNA genome, enclosed within an icosahedral capsid. Viral portal protein plays a key role in the procapsid assembly and DNA packaging. The portal is the entrance and exit pore for the viral genome, making it an attractive pharmacological target for the development of new antivirals. Here we present the atomic structure of the portal protein of Epstein-Barr virus, solved by cryo-electron microscopy at 3.5 Å resolution. The detailed architecture of this protein suggests that it plays a functional role in DNA retention during packaging.


Assuntos
Proteínas do Capsídeo/ultraestrutura , Herpesvirus Humano 4/ultraestrutura , Proteínas Virais/ultraestrutura , Montagem de Vírus , Capsídeo/ultraestrutura , Proteínas do Capsídeo/genética , Microscopia Crioeletrônica , Empacotamento do DNA , DNA Viral/genética , Genoma Viral , Herpesvirus Humano 4/genética , Humanos , Modelos Moleculares , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/ultraestrutura , Proteínas Virais/genética , Vírion/ultraestrutura
18.
Nat Commun ; 10(1): 1456, 2019 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-30926810

RESUMO

Many of the largest known viruses belong to the PRD1-adeno structural lineage characterised by conserved pseudo-hexameric capsomers composed of three copies of a single major capsid protein (MCP). Here, by high-resolution cryo-EM analysis, we show that a class of archaeal viruses possess hetero-hexameric MCPs which mimic the PRD1-adeno lineage trimer. These hetero-hexamers are built from heterodimers and utilise a jigsaw-puzzle system of pegs and holes, and underlying minor capsid proteins, to assemble the capsid laterally from the 5-fold vertices. At these vertices proteins engage inwards with the internal membrane vesicle whilst 2-fold symmetric horn-like structures protrude outwards. The horns are assembled from repeated globular domains attached to a central spine, presumably facilitating multimeric attachment to the cell receptor. Such viruses may represent precursors of the main PRD1-adeno lineage, similarly engaging cell-receptors via 5-fold spikes and using minor proteins to define particle size.


Assuntos
Vírus de Archaea/fisiologia , Montagem de Vírus/fisiologia , Vírus de Archaea/química , Vírus de Archaea/ultraestrutura , Proteínas do Capsídeo/química , Proteínas do Capsídeo/ultraestrutura , Modelos Moleculares
19.
ACS Nano ; 13(4): 4443-4454, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-30912918

RESUMO

As self-assembling polyvalent nanoscale structures that can tolerate substantial genetic and chemical modification, virus-like particles are useful in a variety of fields. Here we describe the genetic modification and structural characterization of the Leviviridae PP7 capsid protein as a platform for the presentation of functional polypeptides. This particle was shown to tolerate the display of sequences from 1 kDa (a cell penetrating peptide) to 14 kDa (the Fc-binding double Z-domain) on its exterior surface as C-terminal genetic fusions to the coat protein. In addition, a dimeric construct allowed the presentation of exogenous loops between capsid monomers and the simultaneous presentation of two different peptides at different positions on the icosahedral structure. The PP7 particle is thereby significantly more tolerant of these types of polypeptide additions than Qß and MS2, the other Leviviridae-derived VLPs in common use.


Assuntos
Proteínas do Capsídeo/química , Capsídeo/química , Leviviridae/química , Biblioteca de Peptídeos , Peptídeos/análise , Sequência de Aminoácidos , Capsídeo/ultraestrutura , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/ultraestrutura , Leviviridae/genética , Leviviridae/ultraestrutura , Modelos Moleculares , Peptídeos/genética , Engenharia de Proteínas
20.
Nat Commun ; 10(1): 1184, 2019 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-30862777

RESUMO

The vertical double ß-barrel major capsid protein (MCP) fold, fingerprint of the PRD1-adeno viral lineage, is widespread in many viruses infecting organisms across the three domains of life. The discovery of PRD1-like viruses with two MCPs challenged the known assembly principles. Here, we present the cryo-electron microscopy (cryo-EM) structures of the archaeal, halophilic, internal membrane-containing Haloarcula californiae icosahedral virus 1 (HCIV-1) and Haloarcula hispanica icosahedral virus 2 (HHIV-2) at 3.7 and 3.8 Å resolution, respectively. Our structures reveal proteins located beneath the morphologically distinct two- and three-tower capsomers and homopentameric membrane proteins at the vertices that orchestrate the positioning of pre-formed vertical single ß-barrel MCP heterodimers. The cryo-EM based structures together with the proteomics data provide insights into the assembly mechanism of this type of viruses and into those with membrane-less double ß-barrel MCPs.


Assuntos
Vírus de Archaea/fisiologia , Proteínas do Capsídeo/ultraestrutura , Vírus de DNA/fisiologia , Haloarcula/virologia , Montagem de Vírus , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Microscopia Crioeletrônica , Modelos Moleculares , Conformação Proteica em Folha beta , Multimerização Proteica , Vírion/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA