Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 219
Filtrar
1.
Viruses ; 14(12)2022 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-36560654

RESUMO

Upon infection, viruses hijack the cell machinery and remodel host cell structures to utilize them for viral proliferation. Since viruses are about a thousand times smaller than their host cells, imaging virus-host interactions at high spatial resolution is like looking for a needle in a haystack. Scouting gross cellular changes with fluorescent microscopy is only possible for well-established viruses, where fluorescent tagging is developed. Soft X-ray tomography (SXT) offers 3D imaging of entire cells without the need for chemical fixation or labeling. Here, we use full-rotation SXT to visualize entire human B cells infected by the herpes simplex virus 1 (HSV-1). We have mapped the temporospatial remodeling of cells during the infection and observed changes in cellular structures, such as the presence of cytoplasmic stress granules and multivesicular structures, formation of nuclear virus-induced dense bodies, and aggregates of capsids. Our results demonstrate the power of SXT imaging for scouting virus-induced changes in infected cells and understanding the orchestration of virus-host remodeling quantitatively.


Assuntos
Herpesvirus Humano 1 , Humanos , Herpesvirus Humano 1/química , Tomografia por Raios X/métodos , Capsídeo
2.
PLoS Pathog ; 18(7): e1010629, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35797345

RESUMO

Herpes simplex virus-1 (HSV-1) is a large, enveloped DNA virus and its assembly in the cell is a complex multi-step process during which viral particles interact with numerous cellular compartments such as the nucleus and organelles of the secretory pathway. Transmission electron microscopy and fluorescence microscopy are commonly used to study HSV-1 infection. However, 2D imaging limits our understanding of the 3D geometric changes to cellular compartments that accompany infection and sample processing can introduce morphological artefacts that complicate interpretation. In this study, we used soft X-ray tomography to observe differences in whole-cell architecture between HSV-1 infected and uninfected cells. To protect the near-native structure of cellular compartments we used a non-disruptive sample preparation technique involving rapid cryopreservation, and a fluorescent reporter virus was used to facilitate correlation of structural changes with the stage of infection in individual cells. We observed viral capsids and assembly intermediates interacting with nuclear and cytoplasmic membranes. Additionally, we observed differences in the morphology of specific organelles between uninfected and infected cells. The local concentration of cytoplasmic vesicles at the juxtanuclear compartment increased and their mean width decreased as infection proceeded, and lipid droplets transiently increased in size. Furthermore, mitochondria in infected cells were elongated and highly branched, suggesting that HSV-1 infection alters the dynamics of mitochondrial fission/fusion. Our results demonstrate that high-resolution 3D images of cellular compartments can be captured in a near-native state using soft X-ray tomography and have revealed that infection causes striking changes to the morphology of intracellular organelles.


Assuntos
Herpes Simples , Herpesvirus Humano 1 , Animais , Núcleo Celular , Chlorocebus aethiops , Herpes Simples/diagnóstico por imagem , Herpesvirus Humano 1/química , Tomografia por Raios X , Células Vero
3.
Nat Nanotechnol ; 16(6): 698-707, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33782587

RESUMO

Expansion microscopy (ExM) physically magnifies biological specimens to enable nanoscale-resolution imaging using conventional microscopes. Current ExM methods permeate specimens with free-radical-chain-growth-polymerized polyacrylate hydrogels, whose network structure limits the local isotropy of expansion as well as the preservation of morphology and shape at the nanoscale. Here we report that ExM is possible using hydrogels that have a more homogeneous network structure, assembled via non-radical terminal linking of tetrahedral monomers. As with earlier forms of ExM, such 'tetra-gel'-embedded specimens can be iteratively expanded for greater physical magnification. Iterative tetra-gel expansion of herpes simplex virus type 1 (HSV-1) virions by ~10× in linear dimension results in a median spatial error of 9.2 nm for localizing the viral envelope layer, rather than 14.3 nm from earlier versions of ExM. Moreover, tetra-gel-based expansion better preserves the virion spherical shape. Thus, tetra-gels may support ExM with reduced spatial errors and improved local isotropy, pointing the way towards single-biomolecule accuracy ExM.


Assuntos
Microscopia/métodos , Polímeros/química , Animais , Encéfalo/citologia , Química Click , Feminino , Células HEK293 , Células HeLa , Herpesvirus Humano 1/química , Humanos , Hidrogéis/química , Processamento de Imagem Assistida por Computador , Masculino , Camundongos Transgênicos , Polietilenoglicóis/química , Polímeros/síntese química , Vírion/ultraestrutura
4.
mBio ; 12(2)2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33653890

RESUMO

Herpes simplex viruses (HSV-1 and HSV-2) encode up to 16 envelope proteins, four of which are essential for entry. However, whether these four proteins alone are sufficient to dictate the broad cellular tropism of HSV-1 and the selection of different cell type-dependent entry routes is unknown. To begin addressing this, we previously pseudotyped vesicular stomatitis virus (VSV), lacking its native glycoprotein G, with only the four essential entry glycoproteins of HSV-1: gB, gH, gL, and gD. This novel VSVΔG-BHLD pseudotype recapitulated several important features of HSV-1 entry: the requirement for gB, gH, gL, gD, and a cellular receptor and sensitivity to anti-gB and anti-gH/gL neutralizing antibodies. However, due to the use of a single cell type in that study, the tropism of the VSVΔG-BHLD pseudotype was not investigated. Here, we show that the cellular tropism of the pseudotype is severely limited compared to that of wild-type HSV-1 and that its entry pathways differ from the native HSV-1 entry pathways. To test the hypothesis that other HSV-1 envelope proteins may contribute to HSV-1 tropism, we generated a derivative pseudotype containing the HSV-1 glycoprotein C (VSVΔG-BHLD-gC) and observed a gC-dependent increase in entry efficiency in two cell types. We propose that the pseudotyping platform developed here has the potential to uncover functional contributions of HSV-1 envelope proteins to entry in a gain-of-function manner.IMPORTANCE Herpes simplex viruses (HSV-1 and HSV-2) contain up to 16 different proteins in their envelopes. Four of these, glycoproteins gB, gD, gH, and gL, are termed essential with regard to entry, whereas the rest are typically referred to as nonessential based on the entry phenotypes of the respective single genetic deletions. However, the single-gene deletion approach, which relies on robust loss-of-function phenotypes, may be confounded by functional redundancies among the many HSV-1 envelope proteins. We have developed a pseudotyping platform in which the essential four entry glycoproteins are isolated from the rest, which can be added back individually for systematic gain-of-function entry experiments. Here, we show the utility of this platform for dissecting the contributions of HSV envelope proteins, both the essential four and the remaining dozen (using gC as an example), to HSV entry.


Assuntos
Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Tipagem Molecular/métodos , Proteínas do Envelope Viral/genética , Tropismo Viral , Internalização do Vírus , Animais , Linhagem Celular , Chlorocebus aethiops , Mutação com Ganho de Função , Células HEK293 , Células HeLa , Herpesvirus Humano 1/química , Humanos , Células Vero , Proteínas do Envelope Viral/metabolismo
5.
PLoS One ; 16(2): e0247396, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33606823

RESUMO

Among various delivery systems for vaccine and drug delivery, cell-penetrating peptides (CPPs) have been known as a potent delivery system because of their capability to penetrate cell membranes and deliver some types of cargoes into cells. Several CPPs were found in the proteome of viruses such as Tat originated from human immunodeficiency virus-1 (HIV-1), and VP22 derived from herpes simplex virus-1 (HSV-1). In the current study, a wide-range of CPPs was identified in the proteome of SARS-CoV-2, a new member of coronaviruses family, using in silico analyses. These CPPs may play a main role for high penetration of virus into cells and infection of host. At first, we submitted the proteome of SARS-CoV-2 to CellPPD web server that resulted in a huge number of CPPs with ten residues in length. Afterward, we submitted the predicted CPPs to C2Pred web server for evaluation of the probability of each peptide. Then, the uptake efficiency of each peptide was investigated using CPPred-RF and MLCPP web servers. Next, the physicochemical properties of the predicted CPPs including net charge, theoretical isoelectric point (pI), amphipathicity, molecular weight, and water solubility were calculated using protparam and pepcalc tools. In addition, the probability of membrane binding potential and cellular localization of each CPP were estimated by Boman index using APD3 web server, D factor, and TMHMM web server. On the other hand, the immunogenicity, toxicity, allergenicity, hemolytic potency, and half-life of CPPs were predicted using various web servers. Finally, the tertiary structure and the helical wheel projection of some CPPs were predicted by PEP-FOLD3 and Heliquest web servers, respectively. These CPPs were divided into: a) CPP containing tumor homing motif (RGD) and/or tumor penetrating motif (RXXR); b) CPP with the highest Boman index; c) CPP with high half-life (~100 hour) in mammalian cells, and d) CPP with +5.00 net charge. Based on the results, we found a large number of novel CPPs with various features. Some of these CPPs possess tumor-specific motifs which can be evaluated in cancer therapy. Furthermore, the novel and potent CPPs derived from SARS-CoV-2 may be used alone or conjugated to some sequences such as nuclear localization sequence (NLS) for vaccine and drug delivery.


Assuntos
Vacinas contra COVID-19/química , Peptídeos Penetradores de Células/química , Biologia Computacional , Simulação por Computador , Sistemas de Liberação de Medicamentos , Proteoma , SARS-CoV-2/química , Animais , COVID-19 , Vacinas contra COVID-19/genética , Vacinas contra COVID-19/metabolismo , Peptídeos Penetradores de Células/genética , Peptídeos Penetradores de Células/metabolismo , HIV-1/química , HIV-1/genética , Herpesvirus Humano 1/química , Herpesvirus Humano 1/genética , Humanos , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/genética , Produtos do Gene tat do Vírus da Imunodeficiência Humana/química , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética
6.
Elife ; 92020 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-32391791

RESUMO

Herpesviruses acquire their membrane envelopes in the cytoplasm of infected cells via a molecular mechanism that remains unclear. Herpes simplex virus (HSV)-1 proteins pUL7 and pUL51 form a complex required for efficient virus envelopment. We show that interaction between homologues of pUL7 and pUL51 is conserved across human herpesviruses, as is their association with trans-Golgi membranes. We characterized the HSV-1 pUL7:pUL51 complex by solution scattering and chemical crosslinking, revealing a 1:2 complex that can form higher-order oligomers in solution, and we solved the crystal structure of the core pUL7:pUL51 heterodimer. While pUL7 adopts a previously-unseen compact fold, the helix-turn-helix conformation of pUL51 resembles the cellular endosomal complex required for transport (ESCRT)-III component CHMP4B and pUL51 forms ESCRT-III-like filaments, suggesting a direct role for pUL51 in promoting membrane scission during virus assembly. Our results provide a structural framework for understanding the role of the conserved pUL7:pUL51 complex in herpesvirus assembly.


Most people suffer from occasional cold sores, which are caused by the herpes simplex virus. This virus causes infections that last your entire life, but for the most part it lies dormant in your cells and reactivates only at times of stress. When it reactivates, the virus manipulates host cells to make new virus particles that may spread the infection to other people. Like many other viruses, herpes simplex viruses also steal jelly-like structures known as membranes from their host cells to form protective coats around new virus particles. In cells from humans and other animals, proteins belonging to a molecular machine known as ESCRT form filaments that bend and break membranes as the cells require. Many viruses hijack the ESCRT machinery to wrap membranes around new virus particles. However, herpes simplex viruses do not follow the usual rules for activating this machine. Instead, they rely on two viral proteins called pUL7 and pUL51 to hot-wire the ESCRT machinery. Previous studies have shown that these two proteins bind to each other, but it remained unclear how they work. Butt et al. used a combination of biochemical and biophysical techniques to solve the three-dimensional structures of pUL7 and pUL51 when bound to each other. The experiments determined that the structure of pUL51 resembles the structures of different components in the ESCRT machinery. Like the ESCRT proteins, pUL51 formed filaments, suggesting that pUL51 bends membranes in cells and that pUL7 blocks it from doing so until the time is right. Further experiments showed that the equivalents of pUL7 and pUL51 in other members of the herpes virus family also bind to each other in a similar way. These findings reveal that herpes simplex viruses and their close relatives have evolved a different strategy than many other viruses to steal membranes from host cells. Interfering with this mechanism may provide new avenues for designing drugs or improving vaccines against these viruses. The pUL7 and pUL51 proteins may also inspire new tools in biotechnology that could precisely control the shapes of biological membranes.


Assuntos
Herpesvirus Humano 1/fisiologia , Fosfoproteínas/química , Fosfoproteínas/genética , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/genética , Proteínas Virais/química , Proteínas Virais/genética , Montagem de Vírus , Células HEK293 , Células HeLa , Herpes Simples/virologia , Herpesvirus Humano 1/química , Humanos , Modelos Moleculares , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Proteínas Virais/metabolismo , Replicação Viral , Rede trans-Golgi
7.
mBio ; 11(3)2020 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-32371601

RESUMO

A distinguishing morphological feature of all herpesviruses is the multiprotein tegument layer located between the nucleocapsid and lipid envelope of the virion. Tegument proteins play multiple roles in viral replication, including viral assembly, but we do not yet understand their individual functions or how the tegument is assembled and organized. UL11, the smallest tegument protein, is important for several distinct processes in replication, including efficient virion morphogenesis and cell-cell spread. However, the mechanistic understanding of its role in these and other processes is limited in part by the scant knowledge of its biochemical and structural properties. Here, we report that UL11 from herpes simplex virus 1 (HSV-1) is an intrinsically disordered, conformationally dynamic protein that undergoes liquid-liquid phase separation (LLPS) in vitro Intrinsic disorder may underlie the ability of UL11 to exert multiple functions and bind multiple partners. Sequence analysis suggests that not only all UL11 homologs but also all HSV-1 tegument proteins contain intrinsically disordered regions of different lengths. The presence of intrinsic disorder, and potentially, the ability to form LLPS, may thus be a common feature of the tegument proteins. We hypothesize that tegument assembly may involve the formation of a biomolecular condensate, driven by the heterogeneous mixture of intrinsically disordered tegument proteins.IMPORTANCE Herpesvirus virions contain a unique tegument layer sandwiched between the capsid and lipid envelope and composed of multiple copies of about two dozen viral proteins. However, little is known about the structure of the tegument or how it is assembled. Here, we show that a conserved tegument protein UL11 from herpes simplex virus 1, a prototypical alphaherpesvirus, is an intrinsically disordered protein that undergoes liquid-liquid phase separation in vitro Through sequence analysis, we find intrinsically disordered regions of different lengths in all HSV-1 tegument proteins. We hypothesize that intrinsic disorder is a common characteristic of tegument proteins and propose a new model of tegument as a biomolecular condensate.


Assuntos
Herpesvirus Humano 1/química , Proteínas de Ligação a RNA/química , Proteínas Estruturais Virais/química , Cristalografia , Herpesvirus Humano 1/genética , Ligação Proteica , Proteínas de Ligação a RNA/genética , Proteínas Estruturais Virais/genética
8.
Methods Mol Biol ; 2060: 279-288, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31617184

RESUMO

The analysis of HSV-1 mature extracellular virions by proteomics requires highly enriched samples to limit false-positives and favor the detection of true components. The protocol described below involves the removal of highly contaminating serum proteins and purification of the virions by a series of differential and density centrifugation steps. In addition, L-particles, which are viral particles devoid of a genome and capsid but present in the extracellular milieu, are depleted on Ficoll 400 gradients. As previously reported, the resulting viral particles are free of most contaminants and suitable for mass spectrometry.


Assuntos
Herpesvirus Humano 1 , Proteômica , Vírion , Células HeLa , Herpesvirus Humano 1/química , Herpesvirus Humano 1/isolamento & purificação , Herpesvirus Humano 1/metabolismo , Humanos , Vírion/química , Vírion/isolamento & purificação , Vírion/metabolismo
9.
Methods Mol Biol ; 2060: 305-317, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31617186

RESUMO

Extracellular vesicles (EVs) are secreted membrane vesicles, derived from endosomes or from the plasma membrane, which have been isolated from most cell types and biological fluids. Although EVs are highly heterogeneous and their classification is complex, two major categories can be distinguished: microvesicles (MVs), which derive from the shedding of the plasma membrane, and exosomes, which correspond to intraluminal vesicles released to the extracellular milieu upon fusion of multivesicular bodies (MVBs) with the plasma membrane. Cells infected with viruses may secrete MVs containing viral proteins, RNAs and, in some instances, infectious virions. A recent study carried out by our laboratory has shown that MVs released by cells infected with HSV-1 contained virions and were endocytosed by naïve cells leading to a productive infection. This suggests that HSV-1 may use MVs for spreading, expanding its tropism and evading the host immune response. Here we describe in detail the methods used to isolate and analyse the MVs released from HSV-1-infected cells.


Assuntos
Micropartículas Derivadas de Células , Herpes Simples/metabolismo , Herpesvirus Humano 1 , Linhagem Celular , Micropartículas Derivadas de Células/química , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/virologia , Herpes Simples/patologia , Herpesvirus Humano 1/química , Herpesvirus Humano 1/isolamento & purificação , Herpesvirus Humano 1/metabolismo , Humanos
10.
Methods Mol Biol ; 2060: 319-326, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31617187

RESUMO

Conformational changes in viral membrane proteins drive membrane fusion, a critical step in virus entry and infection. Here we describe a simple and rapid virus blotting immunoassay to define conformational changes with a panel of monoclonal antibodies to distinct sites across a viral glycoprotein. This dot blot technique has been utilized to define low pH-triggered changes in the prefusion form of the herpesviral fusogen gB. At pH of <6.2 there are specific changes in herpes simplex virus 1 gB domains I and V. This corresponds broadly to host cell endosomal pH. Many of the identified changes are at least partially reversible. This method can be adapted to document changes in viral proteins that are not fusion proteins, including those induced by alternate triggers such as receptor-binding or protease cleavage.


Assuntos
Anticorpos Monoclonais/química , Anticorpos Antivirais/química , Herpesvirus Humano 1 , Immunoblotting , Conformação Proteica , Proteínas do Envelope Viral , Internalização do Vírus , Animais , Chlorocebus aethiops , Herpesvirus Humano 1/química , Herpesvirus Humano 1/metabolismo , Humanos , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo
11.
Methods Mol Biol ; 2060: 377-393, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31617192

RESUMO

Herpes simplex viruses utilize glycoproteins displayed on the viral envelope to perform a variety of functions in the viral infectious cycle. Structural and functional studies of these viral glycoproteins can benefit from biochemical, biophysical, and structural analysis of purified proteins. Here, we describe a general protocol for expression and purification of viral glycoproteins from insect cells based on those developed for the HSV-1 gB and HSV-2 gH/gL ectodomains as well as the protocol for crystallization of these glycoproteins. This protocol can be used for generating milligram amounts of wild-type (WT) or mutant gB and gH/gL ectodomains or can be adapted to produce purified ectodomains of glycoproteins from HSV or other herpesviruses for biochemical and structural studies.


Assuntos
Expressão Gênica , Glicoproteínas , Herpesvirus Humano 1 , Proteínas do Envelope Viral , Animais , Cristalografia por Raios X , Glicoproteínas/biossíntese , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/isolamento & purificação , Herpesvirus Humano 1/química , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Células Sf9 , Spodoptera , Proteínas do Envelope Viral/biossíntese , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/isolamento & purificação
12.
Methods Mol Biol ; 2060: 395-407, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31617193

RESUMO

HSV glycoproteins play important roles in the viral life cycle, particularly viral cell entry. Here we describe the protocol for expression, purification, and crystallization of full-length HSV-1 glycoprotein B. The protocol provides a framework for incorporating transmembrane domain-stabilizing amphipols into the crystallization setup and can be adapted to isolate other complete HSV glycoproteins.


Assuntos
Expressão Gênica , Herpesvirus Humano 1 , Proteínas do Envelope Viral , Animais , Cristalografia por Raios X , Herpesvirus Humano 1/química , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Domínios Proteicos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Células Sf9 , Spodoptera , Proteínas do Envelope Viral/biossíntese , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/isolamento & purificação
13.
Viruses ; 11(9)2019 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-31540043

RESUMO

The herpes simplex virus type 1 (HSV-1) UL37 gene encodes for a multifunctional component of the virion tegument, which is necessary for secondary envelopment in the cytoplasm of infected cells, for motility of the viral particle, and for the first steps in the initiation of virus infection. This 120 kDa protein has several known viral interacting partners, including pUL36, gK/pUL20, pUS10, and VP26, and cellular interacting proteins which include TRAF6, RIG-I, and dystonin. These interactions are likely important for the functions of pUL37 at both early and late stages of infection. We employed a genetic approach to determine essential domains and amino acid residues of pUL37 and their associated functions in cellular localization and virion morphogenesis. Using marker-rescue/marker-transfer methods, we generated a library of GFP-tagged pUL37 mutations in the HSV-1 strain KOS genome. Through viral growth and ultra-structural analysis, we discovered that the C-terminus is essential for replication. The N-terminal 480 amino acids are dispensable for replication in cell culture, although serve some non-essential function as viral titers are reduced in the presence of this truncation. Furthermore, the C-terminal 133 amino acids are important in so much that their absence leads to a lethal phenotype. We further probed the carboxy terminal half of pUL37 by alanine scanning mutagenesis of conserved residues among alphaherpesviruses. Mutant viruses were screened for the inability to form plaques-or greatly reduced plaque size-on Vero cells, of which 22 mutations were chosen for additional analysis. Viruses discovered to have the greatest reduction in viral titers on Vero cells were examined by electron microscopy (EM) and by confocal light microscopy for pUL37-EGFP cellular localization. This genetic approach identified both essential and non-essential domains and residues of the HSV-1 UL37 gene product. The mutations identified in this study are recognized as significant candidates for further analysis of the pUL37 function and may unveil previously undiscovered roles and interactions of this essential tegument gene.


Assuntos
Aminoácidos/genética , Herpesvirus Humano 1/genética , Proteínas Estruturais Virais/genética , Replicação Viral , Aminoácidos/química , Animais , Técnicas de Cultura de Células , Chlorocebus aethiops , Herpesvirus Humano 1/química , Herpesvirus Humano 1/fisiologia , Mutação , Fenótipo , Células Vero , Proteínas Estruturais Virais/química , Vírion/genética , Vírion/metabolismo , Montagem de Vírus
14.
Nature ; 570(7760): 257-261, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31142842

RESUMO

Herpesviruses are enveloped viruses that are prevalent in the human population and are responsible for diverse pathologies, including cold sores, birth defects and cancers. They are characterized by a highly pressurized pseudo-icosahedral capsid-with triangulation number (T) equal to 16-encapsidating a tightly packed double-stranded DNA (dsDNA) genome1-3. A key process in the herpesvirus life cycle involves the recruitment of an ATP-driven terminase to a unique portal vertex to recognize, package and cleave concatemeric dsDNA, ultimately giving rise to a pressurized, genome-containing virion4,5. Although this process has been studied in dsDNA phages6-9-with which herpesviruses bear some similarities-a lack of high-resolution in situ structures of genome-packaging machinery has prevented the elucidation of how these multi-step reactions, which require close coordination among multiple actors, occur in an integrated environment. To better define the structural basis of genome packaging and organization in herpes simplex virus type 1 (HSV-1), we developed sequential localized classification and symmetry relaxation methods to process cryo-electron microscopy (cryo-EM) images of HSV-1 virions, which enabled us to decouple and reconstruct hetero-symmetric and asymmetric elements within the pseudo-icosahedral capsid. Here we present in situ structures of the unique portal vertex, genomic termini and ordered dsDNA coils in the capsid spooled around a disordered dsDNA core. We identify tentacle-like helices and a globular complex capping the portal vertex that is not observed in phages, indicative of herpesvirus-specific adaptations in the DNA-packaging process. Finally, our atomic models of portal vertex elements reveal how the fivefold-related capsid accommodates symmetry mismatch imparted by the dodecameric portal-a longstanding mystery in icosahedral viruses-and inform possible DNA-sequence recognition and headful-sensing pathways involved in genome packaging. This work showcases how to resolve symmetry-mismatched elements in a large eukaryotic virus and provides insights into the mechanisms of herpesvirus genome packaging.


Assuntos
Microscopia Crioeletrônica , Empacotamento do DNA , Genoma Viral , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/ultraestrutura , Conformação de Ácido Nucleico , Capsídeo/química , Capsídeo/ultraestrutura , DNA Viral/química , DNA Viral/ultraestrutura , Herpesvirus Humano 1/química , Modelos Moleculares , Vírion/química , Vírion/genética , Vírion/ultraestrutura
15.
J Immunol Methods ; 465: 31-38, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30502324

RESUMO

We have developed a stable Chinese Hamster Ovary (CHO) cell line for the production of a recombinant monoclonal antibody (mAb) to a short protein sequence derived from the N-terminus of human herpes simplex virus type 1 glycoprotein D (HSV-1 gD). The antibody (designated r34.1) provides a useful tool for the immunoaffinity purification of HSV-1 gD tagged proteins, and provides a generic purification system by which various proteins and peptides can be purified. Recombinant 34.1 was assembled using cDNA derived from a HSV-1 gD specific murine hybridoma engineered to encode a full-length IgG molecule. Antibody expression cassettes were transfected into CHO-S cells, and a stable cell-line expressing up to 500 mg/L of antibody, isolated. Affinity purified r34.1 exhibited nanomolar affinity for its cognate ligand, and is stable throughout multiple cycles of immunoaffinity purification involving ligand binding at neutral pH, followed by acid elution. The HSV-1 gD tag expression and purification strategy has been used to enhance the secretion and purification of several vaccine immunogens including HIV envelope protein rgp120s, but the protocol has potential for generic application.


Assuntos
Anticorpos Monoclonais Murinos/química , Anticorpos Antivirais/química , Herpesvirus Humano 1/química , Proteínas do Envelope Viral/química , Animais , Anticorpos Monoclonais Murinos/genética , Anticorpos Monoclonais Murinos/imunologia , Anticorpos Antivirais/genética , Anticorpos Antivirais/imunologia , Células CHO , Cricetulus , Herpesvirus Humano 1/imunologia , Humanos , Camundongos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas do Envelope Viral/imunologia
16.
Int J Mol Sci ; 19(10)2018 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-30257461

RESUMO

All herpesviruses use a heterodimeric nuclear egress complex (NEC) to transport capsids out of host cell nuclei. Despite their overall similar structure, NECs may differ significantly in sequence between different viruses. Up to now, structural information is limited to isolated NEC heterodimers and to large hexagonal lattices made up of hexagonal ring-like structures ("Hexagons"). The present study aimed to expand the existing structural knowledge with information on the dynamics of NECs from different viruses and in different oligomerization states. For this task, comparative molecular dynamics simulations were performed of the free NEC heterodimers from three different viruses (HCMV (human cytomegalovirus), HSV-1 (herpes simplex virus 1), and PRV (pseudorabies virus)). In addition, higher oligomerization states comprising two or six NEC heterodimers were characterized for HCMV and HSV-1. The study revealed that the isolated NEC heterodimers from α- (HSV-1, PRV) and ß-herpesviruses (HCMV) differ significantly in their dynamics, which can be attributed to a poorly conserved interface region between the NEC subdomains. These differences become smaller for higher oligomerization states, and both HCMV and HSV-1 individual Hexagons exhibit a common region of enhanced dynamics, which might be of functional relevance for the formation of curved vesicle structures or the recognition of hexameric capsid proteins.


Assuntos
Proteínas do Capsídeo/química , Infecções por Herpesviridae/virologia , Herpesviridae/química , Animais , Citomegalovirus/química , Herpesvirus Humano 1/química , Herpesvirus Suídeo 1/química , Humanos , Simulação de Dinâmica Molecular , Conformação Proteica , Multimerização Proteica
17.
J Virol ; 92(18)2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29976665

RESUMO

The herpesvirus capsid assembles in the nucleus as an immature procapsid precursor built around viral scaffold proteins. The event that initiates procapsid maturation is unknown, but it is dependent upon activation of the VP24 internal protease. Scaffold cleavage triggers angularization of the shell and its decoration with the VP26 and pUL25 capsid-surface proteins. In both the procapsid and mature angularized capsid, the apical region of the major capsid protein (VP5) is surface exposed. We investigated whether the VP5 apical region contributes to intracellular transport dynamics following entry into primary sensory neurons and also tested the hypothesis that conserved negatively charged amino acids in the apical region contribute to VP26 acquisition. To our surprise, neither hypothesis proved true. Instead, mutation of glutamic acid residues in the apical region delayed viral propagation and induced focal capsid accumulations in nuclei. Examination of capsid morphogenesis based on epitope unmasking, capsid composition, and ultrastructural analysis indicated that these clusters consisted of procapsids. The results demonstrate that, in addition to established events that occur inside the capsid, the exterior capsid shell promotes capsid morphogenesis and maturation.IMPORTANCE Herpesviruses assemble capsids and encapsidate their genomes by a process that is unlike those of other mammalian viruses but is similar to those of some bacteriophage. Many important aspects of herpesvirus morphogenesis remain enigmatic, including how the capsid shell matures into a stable angularized configuration. Capsid maturation is triggered by activation of a protease that cleaves an internal protein scaffold. We report on the fortuitous discovery that a region of the major capsid protein that is exposed on the outer surface of the capsid also contributes to capsid maturation, demonstrating that the morphogenesis of the capsid shell from its procapsid precursor to the mature angularized form is dependent upon internal and external components of the megastructure.


Assuntos
Proteínas do Capsídeo/genética , Capsídeo/metabolismo , Herpesvirus Humano 1/fisiologia , Proteínas Virais/metabolismo , Animais , Proteínas do Capsídeo/metabolismo , Chlorocebus aethiops , Epitopos/química , Epitopos/genética , Epitopos/metabolismo , Herpesvirus Humano 1/química , Mutação , Células Vero , Proteínas Virais/genética , Vírion/metabolismo , Montagem de Vírus/fisiologia
18.
PLoS Biol ; 16(6): e2006191, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29924793

RESUMO

Herpesviruses include many important human pathogens such as herpes simplex virus, cytomegalovirus, varicella-zoster virus, and the oncogenic Epstein-Barr virus and Kaposi sarcoma-associated herpesvirus. Herpes virions contain a large icosahedral capsid that has a portal at a unique 5-fold vertex, similar to that seen in the tailed bacteriophages. The portal is a molecular motor through which the viral genome enters the capsid during virion morphogenesis. The genome also exits the capsid through the portal-vertex when it is injected through the nuclear pore into the nucleus of a new host cell to initiate infection. Structural investigations of the herpesvirus portal-vertex have proven challenging, owing to the small size of the tail-like portal-vertex-associated tegument (PVAT) and the presence of the tegument layer that lays between the nucleocapsid and the viral envelope, obscuring the view of the portal-vertex. Here, we show the structure of the herpes simplex virus portal-vertex at subnanometer resolution, solved by electron cryomicroscopy (cryoEM) and single-particle 3D reconstruction. This led to a number of new discoveries, including the presence of two previously unknown portal-associated structures that occupy the sites normally taken by the penton and the Ta triplex. Our data revealed that the PVAT is composed of 10 copies of the C-terminal domain of pUL25, which are uniquely arranged as two tiers of star-shaped density. Our 3D reconstruction of the portal-vertex also shows that one end of the viral genome extends outside the portal in the manner described for some bacteriophages but not previously seen in any eukaryote viruses. Finally, we show that the viral genome is consistently packed in a highly ordered left-handed spool to form concentric shells of DNA. Our data provide new insights into the structure of a molecular machine critical to the biology of an important class of human pathogens.


Assuntos
Capsídeo/ultraestrutura , Herpesvirus Humano 1/ultraestrutura , Capsídeo/química , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/ultraestrutura , Microscopia Crioeletrônica , DNA Viral/química , Genoma Viral , Herpesvirus Humano 1/química , Herpesvirus Humano 1/genética , Humanos , Imageamento Tridimensional , Modelos Biológicos , Modelos Moleculares , Proteínas Motores Moleculares/química , Proteínas Motores Moleculares/genética , Proteínas Motores Moleculares/ultraestrutura , Montagem de Vírus
19.
Science ; 360(6384)2018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29622628

RESUMO

Herpes simplex viruses (HSVs) rely on capsid-associated tegument complex (CATC) for long-range axonal transport of their genome-containing capsids between sites of infection and neuronal cell bodies. Here we report cryo-electron microscopy structures of the HSV-1 capsid with CATC up to 3.5-angstrom resolution and atomic models of multiple conformers of capsid proteins VP5, VP19c, VP23, and VP26 and tegument proteins pUL17, pUL25, and pUL36. Crowning every capsid vertex are five copies of heteropentameric CATC, each containing a pUL17 monomer supporting the coiled-coil helix bundle of a pUL25 dimer and a pUL36 dimer, thus positioning their flexible domains for potential involvement in nuclear capsid egress and axonal capsid transport. Notwithstanding newly discovered fold conservation between triplex proteins and bacteriophage λ protein gpD and the previously recognized bacteriophage HK97 gp5-like fold in VP5, HSV-1 capsid proteins exhibit extraordinary diversity in forms of domain insertion and conformational polymorphism, not only for interactions with tegument proteins but also for encapsulation of large genomes.


Assuntos
Proteínas do Capsídeo/química , Capsídeo/química , Herpesvirus Humano 1/química , Animais , Capsídeo/ultraestrutura , Proteínas do Capsídeo/ultraestrutura , Chlorocebus aethiops , Microscopia Crioeletrônica , Glicoproteínas/química , Glicoproteínas/ultraestrutura , Herpesvirus Humano 1/ultraestrutura , Humanos , Conformação Proteica em alfa-Hélice , Células Vero
20.
J Virol ; 92(11)2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29514910

RESUMO

Herpes simplex virus 1 (HSV-1) establishes a lifelong latent infection in host peripheral neurons, including the neurons of the trigeminal ganglia (TG). HSV-1 can reactivate from neurons to cause recurrent infection. During latency, the insulator protein CTCF occupies DNA binding sites on the HSV-1 genome, and these sites have been previously characterized as functional enhancer-blocking insulators. Previously, CTCF was found to be dissociated from wild-type virus postreactivation but not in mutants that do not reactivate, indicating that CTCF eviction may also be an important component of reactivation. To further elucidate the role of CTCF in reactivation of HSV-1, we used recombinant adeno-associated virus (rAAV) vectors to deliver a small interfering RNA targeting CTCF to peripheral neurons latent with HSV-1 in rabbit TG. Our data show that CTCF depletion resulted in long-term and persistent shedding of infectious virus in the cornea and increased ICP0 expression in the ganglia, indicating that CTCF depletion facilitates HSV-1 reactivation.IMPORTANCE Increasing evidence has shown that the insulator protein CTCF regulates gene expression of DNA viruses, including the gammaherpesviruses. While CTCF occupation and insulator function control gene expression in DNA viruses, CTCF eviction has been correlated to increased lytic gene expression and the dissolution of transcriptional domains. Our previous data have shown that in the alphaherpesvirus HSV-1, CTCF was found to be dissociated from the HSV-1 genome postreactivation, further indicating a global role for CTCF eviction in the transition from latency to reactivation in HSV-1 genomes. Using an rAAV8, we targeted HSV-1-infected peripheral neurons for CTCF depletion to show that CTCF depletion precedes the shedding of infectious virus and increased lytic gene expression in vivo, providing the first evidence that CTCF depletion facilitates HSV-1 reactivation.


Assuntos
Fator de Ligação a CCCTC/genética , Técnicas de Inativação de Genes/métodos , Herpes Simples/genética , Herpesvirus Humano 1/fisiologia , Células 3T3 , Animais , Sítios de Ligação , Fator de Ligação a CCCTC/metabolismo , Córnea/virologia , Modelos Animais de Doenças , Gânglios/virologia , Genoma Viral , Herpes Simples/virologia , Herpesvirus Humano 1/química , Camundongos , Coelhos , Ativação Viral , Latência Viral , Eliminação de Partículas Virais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...