Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Curr Top Microbiol Immunol ; 438: 25-58, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-34731265

RESUMO

The virions of all enveloped viruses, including those of the Herpesviridae, must bind to the cell surface then undergo a process of membrane fusion between the cell plasma membrane and the virus particle envelope. As for all herpesviruses, glycoproteins in the virion envelope are the modus operandi of these events.


Assuntos
Herpesviridae , Proteínas do Envelope Viral , Proteínas do Envelope Viral/metabolismo , Glicoproteínas/metabolismo , Vírion/metabolismo , Membrana Celular/metabolismo
2.
PLoS Pathog ; 17(1): e1008961, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33411789

RESUMO

Varicella-zoster virus (VZV) is a medically important alphaherpesvirus that induces fusion of the virion envelope and the cell membrane during entry, and between cells to form polykaryocytes within infected tissues during pathogenesis. All members of the Herpesviridae, including VZV, have a conserved core fusion complex composed of glycoproteins, gB, gH and gL. The ectodomain of the primary fusogen, gB, has five domains, DI-V, of which DI contains the fusion loops needed for fusion function. We recently demonstrated that DIV is critical for fusion initiation, which was revealed by a 2.8Å structure of a VZV neutralizing mAb, 93k, bound to gB and mutagenesis of the gB-93k interface. To further assess the mechanism of mAb 93k neutralization, the binding site of a non-neutralizing mAb to gB, SG2, was compared to mAb 93k using single particle cryogenic electron microscopy (cryo-EM). The gB-SG2 interface partially overlapped with that of gB-93k but, unlike mAb 93k, mAb SG2 did not interact with the gB N-terminus, suggesting a potential role for the gB N-terminus in membrane fusion. The gB ectodomain structure in the absence of antibody was defined at near atomic resolution by single particle cryo-EM (3.9Å) of native, full-length gB purified from infected cells and by X-ray crystallography (2.4Å) of the transiently expressed ectodomain. Both structures revealed that the VZV gB N-terminus (aa72-114) was flexible based on the absence of visible structures in the cryo-EM or X-ray crystallography data but the presence of gB N-terminal peptides were confirmed by mass spectrometry. Notably, N-terminal residues 109KSQD112 were predicted to form a small α-helix and alanine substitution of these residues abolished cell-cell fusion in a virus-free assay. Importantly, transferring the 109AAAA112 mutation into the VZV genome significantly impaired viral propagation. These data establish a functional role for the gB N-terminus in membrane fusion broadly relevant to the Herpesviridae.


Assuntos
Herpesvirus Humano 3/fisiologia , Melanoma/metabolismo , Fusão de Membrana , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Sequência de Aminoácidos , Cristalografia por Raios X , Humanos , Melanoma/virologia , Modelos Moleculares , Mutação , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Homologia de Sequência , Células Tumorais Cultivadas , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
3.
PLoS Pathog ; 16(11): e1009022, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33216797

RESUMO

Cell-cell fusion (abbreviated as cell fusion) is a characteristic pathology of medically important viruses, including varicella-zoster virus (VZV), the causative agent of chickenpox and shingles. Cell fusion is mediated by a complex of VZV glycoproteins, gB and gH-gL, and must be tightly regulated to enable skin pathogenesis based on studies with gB and gH hyperfusogenic VZV mutants. Although the function of gB and gH-gL in the regulation of cell fusion has been explored, whether host factors are directly involved in this regulation process is unknown. Here, we discovered host factors that modulated VZV gB/gH-gL mediated cell fusion via high-throughput screening of bioactive compounds with known cellular targets. Two structurally related non-antibiotic macrolides, tacrolimus and pimecrolimus, both significantly increased VZV gB/gH-gL mediated cell fusion. These compounds form a drug-protein complex with FKBP1A, which binds to calcineurin and specifically inhibits calcineurin phosphatase activity. Inhibition of calcineurin phosphatase activity also enhanced both herpes simplex virus-1 fusion complex and syncytin-1 mediated cell fusion, indicating a broad role of calcineurin in modulating this process. To characterize the role of calcineurin phosphatase activity in VZV gB/gH-gL mediated fusion, a series of biochemical, biological and infectivity assays was performed. Pimecrolimus-induced, enhanced cell fusion was significantly reduced by shRNA knockdown of FKBP1A, further supporting the role of calcineurin phosphatase activity in fusion regulation. Importantly, inhibition of calcineurin phosphatase activity during VZV infection caused exaggerated syncytia formation and suppressed virus propagation, which was consistent with the previously reported phenotypes of gB and gH hyperfusogenic VZV mutants. Seven host cell proteins that remained uniquely phosphorylated when calcineurin phosphatase activity was inhibited were identified as potential downstream factors involved in fusion regulation. These findings demonstrate that calcineurin is a critical host cell factor pivotal in the regulation of VZV induced cell fusion, which is essential for VZV pathogenesis.


Assuntos
Calcineurina/metabolismo , Varicela/virologia , Herpes Zoster/virologia , Herpesvirus Humano 3/fisiologia , Glicoproteínas de Membrana/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/metabolismo , Fusão Celular , Linhagem Celular , Glicoproteínas/metabolismo , Herpesvirus Humano 3/genética , Humanos , Mutação
4.
Proteins ; 89(12): 1647-1672, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34561912

RESUMO

The biological and functional significance of selected Critical Assessment of Techniques for Protein Structure Prediction 14 (CASP14) targets are described by the authors of the structures. The authors highlight the most relevant features of the target proteins and discuss how well these features were reproduced in the respective submitted predictions. The overall ability to predict three-dimensional structures of proteins has improved remarkably in CASP14, and many difficult targets were modeled with impressive accuracy. For the first time in the history of CASP, the experimentalists not only highlighted that computational models can accurately reproduce the most critical structural features observed in their targets, but also envisaged that models could serve as a guidance for further studies of biologically-relevant properties of proteins.


Assuntos
Modelos Moleculares , Conformação Proteica , Proteínas/química , Software , Sequência de Aminoácidos , Biologia Computacional , Microscopia Crioeletrônica , Cristalografia por Raios X , Análise de Sequência de Proteína
5.
Biochem Soc Trans ; 48(6): 2415-2435, 2020 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-33259590

RESUMO

Varicella-zoster virus (VZV) is the causative agent of chicken pox (varicella) and shingles (zoster). Although considered benign diseases, both varicella and zoster can cause complications. Zoster is painful and can lead to post herpetic neuralgia. VZV has also been linked to stroke, related to giant cell arteritis in some cases. Vaccines are available but the attenuated vaccine is not recommended in immunocompromised individuals and the efficacy of the glycoprotein E (gE) based subunit vaccine has not been evaluated for the prevention of varicella. A hallmark of VZV pathology is the formation of multinucleated cells termed polykaryocytes in skin lesions. This cell-cell fusion (abbreviated as cell fusion) is mediated by the VZV glycoproteins gB, gH and gL, which constitute the fusion complex of VZV, also needed for virion entry. Expression of gB, gH and gL during VZV infection and trafficking to the cell surface enables cell fusion. Recent evidence supports the concept that cellular processes are required for regulating cell fusion induced by gB/gH-gL. Mutations within the carboxyl domains of either gB or gH have profound effects on fusion regulation and dramatically restrict the ability of VZV to replicate in human skin. This loss of regulation modifies the transcriptome of VZV infected cells. Furthermore, cellular proteins have significant effects on the regulation of gB/gH-gL-mediated cell fusion and the replication of VZV, exemplified by the cellular phosphatase, calcineurin. This review provides the current state-of-the-art knowledge about the molecular controls of cell fusion-dependent pathogenesis caused by VZV.


Assuntos
Herpesvirus Humano 3/imunologia , Interações Hospedeiro-Patógeno , Infecção pelo Vírus da Varicela-Zoster/virologia , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Animais , Fusão Celular , Vacina contra Varicela , Dimerização , Regulação Viral da Expressão Gênica , Herpesvirus Humano 3/genética , Humanos , Imunoglobulina E/química , Glicoproteínas de Membrana/metabolismo , Camundongos , Mutagênese , Mutação , Fases de Leitura Aberta , Conformação Proteica , Infecção pelo Vírus da Varicela-Zoster/imunologia , Infecção pelo Vírus da Varicela-Zoster/prevenção & controle , Proteínas Virais/metabolismo , Vírion/metabolismo
6.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27795423

RESUMO

The highly conserved herpesvirus glycoprotein complex gB/gH-gL mediates membrane fusion during virion entry and cell-cell fusion. Varicella-zoster virus (VZV) characteristically forms multinucleated cells, or syncytia, during the infection of human tissues, but little is known about this process. The cytoplasmic domain of VZV gB (gBcyt) has been implicated in cell-cell fusion regulation because a gB[Y881F] substitution causes hyperfusion. gBcyt regulation is necessary for VZV pathogenesis, as the hyperfusogenic mutant gB[Y881F] is severely attenuated in human skin xenografts. In this study, gBcyt-regulated fusion was investigated by comparing melanoma cells infected with wild-type-like VZV or hyperfusogenic mutants. The gB[Y881F] mutant exhibited dramatically accelerated syncytium formation in melanoma cells caused by fusion of infected cells with many uninfected cells, increased cytoskeleton reorganization, and rapid displacement of nuclei to dense central structures compared to pOka using live-cell confocal microscopy. VZV and human transcriptomes were concurrently investigated using whole transcriptome sequencing (RNA-seq) to identify viral and cellular responses induced when gBcyt regulation was disrupted by the gB[Y881F] substitution. The expression of four vital VZV genes, ORF61 and the genes for glycoproteins gC, gE, and gI, was significantly reduced at 36 h postinfection for the hyperfusogenic mutants. Importantly, hierarchical clustering demonstrated an association of differential gene expression with dysregulated gBcyt-mediated fusion. A subset of Ras GTPase genes linked to membrane remodeling were upregulated in cells infected with the hyperfusogenic mutants. These data implicate gBcyt in the regulation of gB fusion function that, if unmodulated, triggers cellular processes leading to hyperfusion that attenuates VZV infection. IMPORTANCE: The highly infectious, human-restricted pathogen varicella-zoster virus (VZV) causes chickenpox and shingles. Postherpetic neuralgia (PHN) is a common complication of shingles that manifests as prolonged excruciating pain, which has proven difficult to treat. The formation of fused multinucleated cells in ganglia might be associated with this condition. An effective vaccine against VZV is available but not recommended for immunocompromised individuals, highlighting the need for new therapies. This study investigated the viral and cellular responses to hyperfusion, a condition where the usual constraints of cell membranes are overcome and cells form multinucleated cells. This process hinders VZV and is regulated by a viral glycoprotein, gB. A combination of live-cell imaging and next-generation genomics revealed an alteration in viral and cellular responses during hyperfusion that was caused by the loss of gB regulation. These studies reveal mechanisms central to VZV pathogenesis, potentially leading to improved therapies.


Assuntos
Herpesvirus Humano 3/genética , Interações Hospedeiro-Patógeno , Melanócitos/virologia , Transcriptoma , Proteínas do Envelope Viral/genética , Proteínas ras/genética , Substituição de Aminoácidos , Fusão Celular , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Ontologia Genética , Genes Reporter , Células Gigantes/imunologia , Células Gigantes/ultraestrutura , Células Gigantes/virologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Herpesvirus Humano 3/crescimento & desenvolvimento , Herpesvirus Humano 3/imunologia , Humanos , Melanócitos/imunologia , Melanócitos/ultraestrutura , Anotação de Sequência Molecular , Mutação , Domínios Proteicos , Análise de Sequência de RNA , Transdução de Sinais , Proteínas do Envelope Viral/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia , Internalização do Vírus , Proteínas ras/imunologia
7.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27795427

RESUMO

The conserved glycoproteins gB and gH-gL are essential for herpesvirus entry and cell-cell fusion induced syncytium formation, a characteristic of varicella-zoster virus (VZV) pathology in skin and sensory ganglia. VZV syncytium formation, which has been implicated in the painful condition of postherpetic neuralgia, is regulated by the cytoplasmic domains of gB (gBcyt) via an immunoreceptor tyrosine-based inhibition motif (ITIM) and gH (gHcyt). A lysine cluster (K894, K897, K898, and K900) in the VZV gBcyt was identified by sequence alignment to be conserved among alphaherpesviruses, suggesting a functional role. Alanine and arginine substitutions were used to determine if the positive charge and susceptibility to posttranslational modifications of these lysines contributed to gB/gH-gL cell-cell fusion. Critically, the positive charge of the lysine residues was necessary for fusion regulation, as alanine substitutions induced a 440% increase in fusion compared to that of the wild-type gBcyt while arginine substitutions had wild-type-like fusion levels in an in vitro gB/gH-gL cell fusion assay. Consistent with these results, the alanine substitutions in the viral genome caused exaggerated syncytium formation, reduced VZV titers (-1.5 log10), and smaller plaques than with the parental Oka (pOka) strain. In contrast, arginine substitutions resulted in syncytia with only 2-fold more nuclei, a -0.5-log10 reduction in titers, and pOka-like plaques. VZV mutants with both an ITIM mutation and either alanine or arginine substitutions had reduced titers and small plaques but differed in syncytium morphology. Thus, effective VZV propagation is dependent on cell-cell fusion regulation by the conserved gBcyt lysine cluster, in addition to the gBcyt ITIM and the gHcyt. IMPORTANCE: Varicella-zoster virus (VZV) is a ubiquitous pathogen that causes chickenpox and shingles. Individuals afflicted with shingles risk developing the painful condition of postherpetic neuralgia (PHN), which has been difficult to treat because the underlying cause is not well understood. Additional therapies are needed, as the current vaccine is not recommended for immunocompromised individuals and its efficacy decreases with the age of the recipient. VZV is known to induce the formation of multinuclear cells in neuronal tissue, which has been proposed to be a factor contributing to PHN. This study examines the role of a lysine cluster in the cytoplasmic domain of the VZV fusion protein, gB, in the formation of VZV induced multinuclear cells and in virus replication kinetics and spread. The findings further elucidate how VZV self-regulates multinuclear cell formation and may provide insight into the development of new PHN therapies.


Assuntos
Células Epiteliais/virologia , Herpesvirus Humano 3/crescimento & desenvolvimento , Lisina/química , Glicoproteínas de Membrana/química , Processamento de Proteína Pós-Traducional , Proteínas do Envelope Viral/química , Proteínas Virais/química , Alanina/química , Alanina/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Arginina/química , Arginina/metabolismo , Células CHO , Fusão Celular , Linhagem Celular Tumoral , Sequência Conservada , Cricetulus , Células Epiteliais/imunologia , Expressão Gênica , Células Gigantes/ultraestrutura , Células Gigantes/virologia , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/imunologia , Interações Hospedeiro-Patógeno , Humanos , Lisina/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Mutação , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Alinhamento de Sequência , Eletricidade Estática , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia , Internalização do Vírus
8.
Proc Natl Acad Sci U S A ; 112(19): 6056-61, 2015 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-25918416

RESUMO

Varicella-zoster virus (VZV), of the family Alphaherpesvirinae, causes varicella in children and young adults, potentially leading to herpes zoster later in life on reactivation from latency. The conserved herpesvirus glycoprotein gB and the heterodimer gHgL mediate virion envelope fusion with cell membranes during virus entry. Naturally occurring neutralizing antibodies against herpesviruses target these entry proteins. To determine the molecular basis for VZV neutralization, crystal structures of gHgL were determined in complex with fragments of antigen binding (Fabs) from two human monoclonal antibodies, IgG-94 and IgG-RC, isolated from seropositive subjects. These structures reveal that the antibodies target the same site, composed of residues from both gH and gL, distinct from two other neutralizing epitopes identified by negative-stain electron microscopy and mutational analysis. Inhibition of gB/gHgL-mediated membrane fusion and structural comparisons with herpesvirus homologs suggest that the IgG-RC/94 epitope is in proximity to the site on VZV gHgL that activates gB. Immunization studies proved that the anti-gHgL IgG-RC/94 epitope is a critical target for antibodies that neutralize VZV. Thus, the gHgL/Fab structures delineate a site of herpesvirus vulnerability targeted by natural immunity.


Assuntos
Anticorpos Neutralizantes/química , Glicoproteínas/química , Herpesvirus Humano 3/imunologia , Proteínas do Envelope Viral/química , Animais , Anticorpos Monoclonais/química , Anticorpos Antivirais/imunologia , Cristalografia por Raios X , Epitopos/química , Humanos , Fragmentos de Imunoglobulinas/química , Camundongos , Modelos Moleculares , Testes de Neutralização , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ressonância de Plasmônio de Superfície
9.
J Virol ; 90(16): 7567-78, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27279620

RESUMO

UNLABELLED: Varicella-zoster virus (VZV) is an alphaherpesvirus that causes varicella and herpes zoster. Membrane fusion is essential for VZV entry and the distinctive syncytium formation in VZV-infected skin and neuronal tissue. Herpesvirus fusion is mediated by a complex of glycoproteins gB and gH-gL, which are necessary and sufficient for VZV to induce membrane fusion. However, the cellular requirements of fusion are poorly understood. Integrins have been implicated to facilitate entry of several human herpesviruses, but their role in VZV entry has not yet been explored. To determine the involvement of integrins in VZV fusion, a quantitative cell-cell fusion assay was developed using a VZV-permissive melanoma cell line. The cells constitutively expressed a reporter protein and short hairpin RNAs (shRNAs) to knock down the expression of integrin subunits shown to be expressed in these cells by RNA sequencing. The αV integrin subunit was identified as mediating VZV gB/gH-gL fusion, as its knockdown by shRNAs reduced fusion levels to 60% of that of control cells. A comparable reduction in fusion levels was observed when an anti-αV antibody specific to its extracellular domain was tested in the fusion assay, confirming that the domain was important for VZV fusion. In addition, reduced spread was observed in αV knockdown cells infected with the VZV pOka strain relative to that of the control cells. This was demonstrated by reductions in plaque size, replication kinetics, and virion entry in the αV subunit knockdown cells. Thus, the αV integrin subunit is important for VZV gB/gH-gL fusion and infection. IMPORTANCE: Varicella-zoster virus (VZV) is a highly infectious pathogen that causes chickenpox and shingles. A common complication of shingles is the excruciating condition called postherpetic neuralgia, which has proven difficult to treat. While a vaccine is now available, it is not recommended for immunocompromised individuals and its efficacy decreases with the recipient's age. These limitations highlight the need for new therapies. This study examines the role of integrins in membrane fusion mediated by VZV glycoproteins gB and gH-gL, a required process for VZV infection. This knowledge will further the understanding of VZV entry and provide insight into the development of better therapies.


Assuntos
Herpesvirus Humano 3/fisiologia , Interações Hospedeiro-Patógeno , Integrina alfaV/metabolismo , Internalização do Vírus , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Integrina alfaV/genética , Ensaio de Placa Viral
10.
PLoS Pathog ; 10(5): e1004173, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24874654

RESUMO

The conserved herpesvirus fusion complex consists of glycoproteins gB, gH, and gL which is critical for virion envelope fusion with the cell membrane during entry. For Varicella Zoster Virus (VZV), the complex is necessary for cell-cell fusion and presumed to mediate entry. VZV causes syncytia formation via cell-cell fusion in skin and in sensory ganglia during VZV reactivation, leading to neuronal damage, a potential contributory factor for the debilitating condition of postherpetic neuralgia. The gH cytoplasmic domain (gHcyt) is linked to the regulation of gB/gH-gL-mediated cell fusion as demonstrated by increased cell fusion in vitro by an eight amino acid (aa834-841) truncation of the gHcyt. The gHcyt regulation was identified to be dependent on the physical presence of the domain, and not of specific motifs or biochemical properties as substitution of aa834-841 with V5, cMyc, and hydrophobic or hydrophilic sequences did not affect fusion. The importance of the gHcyt length was corroborated by stepwise deletions of aa834-841 causing incremental increases in cell fusion, independent of gH surface expression and endocytosis. Consistent with the fusion assay, truncating the gHcyt in the viral genome caused exaggerated syncytia formation and significant reduction in viral titers. Importantly, infection of human skin xenografts in SCID mice was severely impaired by the truncation while maintaining the gHcyt length with the V5 substitution preserved typical replication in vitro and in skin. A role for the gHcyt in modulating the functions of the gB cytoplasmic domain (gBcyt) is proposed as the gHcyt truncation substantially enhanced cell fusion in the presence of the gB[Y881F] mutation. The significant reduction in skin infection caused by hyperfusogenic mutations in either the gHcyt or gBcyt demonstrates that both domains are critical for regulating syncytia formation and failure to control cell fusion, rather than enhancing viral spread, is severely detrimental to VZV pathogenesis.


Assuntos
Herpesvirus Humano 3/metabolismo , Glicoproteínas de Membrana/metabolismo , Pele/imunologia , Proteínas Virais/metabolismo , Replicação Viral/fisiologia , Animais , Fusão Celular , Humanos , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos SCID , Pele/metabolismo , Pele/virologia , Proteínas Virais/imunologia , Vírion/metabolismo , Replicação Viral/imunologia
11.
Proc Natl Acad Sci U S A ; 110(5): 1911-6, 2013 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-23322733

RESUMO

Herpesvirus entry functions of the conserved glycoproteins gB and gH-gL have been delineated, but their role in regulating cell-cell fusion is poorly understood. Varicella-zoster virus (VZV) infection provides a valuable model for investigating cell-cell fusion because of the importance of this process for pathogenesis in human skin and sensory ganglia. The present study identifies a canonical immunoreceptor tyrosine-based inhibition motif (ITIM) in the gB cytoplasmic domain (gBcyt) and demonstrates that the gBcyt is a tyrosine kinase substrate. Orbitrap mass spectrometry confirmed that Y881, central to the ITIM, is phosphorylated. To determine whether the gBcyt ITIM regulates gB/gH-gL-induced cell-cell fusion in vitro, tyrosine residues Y881 and Y920 in the gBcyt were substituted with phenylalanine separately or together. Recombinant viruses with these substitutions were generated to establish their effects on syncytia formation in replication in vitro and in the human skin xenograft model of VZV pathogenesis. The Y881F substitution caused significantly increased cell-cell fusion despite reduced cell-surface gB. Importantly, the Y881F or Y881/920F substitutions in VZV caused aggressive syncytia formation, reducing cell-cell spread. These in vitro effects of aggressive syncytia formation translated to severely impaired skin infection in vivo. In contrast, the Y920F substitution did not affect virus replication in vitro or in vivo. These observations suggest that gB modulates cell-cell fusion via an ITIM-mediated Y881 phosphorylation-dependent mechanism, supporting a unique concept that intracellular signaling through this gBcyt motif regulates VZV syncytia formation and is essential for skin pathogenesis.


Assuntos
Herpesvirus Humano 3/metabolismo , Motivo de Inibição do Imunorreceptor Baseado em Tirosina , Pele/patologia , Proteínas do Envelope Viral/metabolismo , Sequência de Aminoácidos , Animais , Western Blotting , Células CHO , Fusão Celular , Linhagem Celular Tumoral , Células Cultivadas , Cricetinae , Cricetulus , Células Gigantes/ultraestrutura , Células Gigantes/virologia , Células HEK293 , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/fisiologia , Humanos , Melanoma/patologia , Melanoma/ultraestrutura , Melanoma/virologia , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Modelos Moleculares , Mutação , Fosforilação , Estrutura Terciária de Proteína , Pele/virologia , Transplante Heterólogo , Tirosina/genética , Tirosina/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
12.
J Virol ; 87(9): 5106-17, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23427162

RESUMO

The tegument proteins encoded by ORF11 and ORF9 of varicella-zoster virus (VZV) are conserved among all alphaherpesvirus. We previously demonstrated that the ORF9 gene is essential, whereas ORF11 is dispensable in vitro but its deletion severely impairs VZV infection of skin xenografts in the SCID mouse model in vivo. Here we report that ORF11 protein interacts with ORF9 protein in infected cells as well as in the absence of other viral proteins, and we have mapped the ORF11 protein domain involved in their interaction. Although ORF11 is an RNA binding protein, the interaction between ORF11 and ORF9 proteins was not mediated by RNA or DNA bridging. VZV recombinants with mutations preventing ORF11 protein binding to ORF9 protein had no effect on 6-day growth kinetics based on plaque numbers, but plaque sizes were reduced in vitro. However, disruption of the ORF11 and ORF9 protein interaction was associated with failure to replicate in skin xenografts in vivo. Further, we demonstrate that in the absence of their interaction, the ORF9 protein displays an identical cellular localization, accumulating in the trans-Golgi region, whereas the ORF11 protein exhibits aberrant localization, dispersing throughout the cytoplasm. Overall, our observations suggest that while complete tegument assembly may not be necessary for VZV replication in vitro, the interaction between the ORF11 and ORF9 proteins appears to be critical for the proper localization of ORF11 protein to the assembly complex and for production of infectious virus during VZV pathogenesis in skin.


Assuntos
Varicela/virologia , Herpesvirus Humano 3/metabolismo , Fases de Leitura Aberta , Proteínas de Ligação a RNA/metabolismo , Proteínas Estruturais Virais/metabolismo , Animais , Linhagem Celular , Herpesvirus Humano 3/genética , Humanos , Camundongos , Camundongos SCID , Ligação Proteica , Proteínas de Ligação a RNA/genética , Proteínas Estruturais Virais/genética
13.
Proc Natl Acad Sci U S A ; 108(45): 18412-7, 2011 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-22025718

RESUMO

Enveloped viruses require membrane fusion for cell entry and replication. For herpesviruses, this event is governed by the multiprotein core complex of conserved glycoproteins (g)B and gH/gL. The recent crystal structures of gH/gL from herpes simplex virus 2, pseudorabies virus, and Epstein-Barr virus revealed distinct domains that, surprisingly, do not resemble known viral fusogens. Varicella-zoster virus (VZV) causes chicken pox and shingles. VZV is an α-herpesvirus closely related to herpes simplex virus 2, enabling prediction of the VZV gH structure by homology modeling. We have defined specific roles for each gH domain in VZV replication and pathogenesis using structure-based site-directed mutagenesis of gH. The distal tip of domain (D)I was important for skin tropism, entry, and fusion. DII helices and a conserved disulfide bond were essential for gH structure and VZV replication. An essential (724)CXXC(727) motif was critical for DIII structural stability and membrane fusion. This assignment of domain-dependent mechanisms to VZV gH links elements of the glycoprotein structure to function in herpesvirus replication and virulence.


Assuntos
Herpesvirus Humano 3/fisiologia , Fusão de Membrana/fisiologia , Pele/virologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/fisiologia , Tropismo Viral/fisiologia , Herpesvirus Humano 3/patogenicidade , Modelos Moleculares , Mutagênese Sítio-Dirigida , Relação Estrutura-Atividade , Proteínas do Envelope Viral/genética , Virulência , Replicação Viral
14.
bioRxiv ; 2024 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-37577490

RESUMO

Cryogenic-electron tomography (cryo-ET) has provided an un-precedented glimpse into the nanoscale architecture of cells by combining cryogenic preservation of biological structures with electron tomography. Micropatterning of extracellular matrix proteins is increasingly used as a method to prepare adherent cell types for cryo-ET as it promotes optimal positioning of cells and subcellular regions of interest for vitrification, cryo-focused ion beam (cryo-FIB) milling, and data acquisition. Here we demonstrate a micropatterning workflow for capturing minimally adherent cell types, human T-cells and Jurkat cells, for cryo-FIB and cryo-ET. Our affinity capture system facilitated the nanoscale imaging of Jurkat cells, revealing extracellular filamentous structures. It improved workflow efficiency by consistently producing grids with a sufficient number of well-positioned cells for an entire cryo-FIB session. Affinity capture can be extended to facilitate high resolution imaging of other adherent and non-adherent cell types with cryo-ET.

15.
PLoS Pathog ; 7(8): e1002157, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21901090

RESUMO

Promyelocytic leukemia protein (PML) has antiviral functions and many viruses encode gene products that disrupt PML nuclear bodies (PML NBs). However, evidence of the relevance of PML NB modification for viral pathogenesis is limited and little is known about viral gene functions required for PML NB disruption in infected cells in vivo. Varicella-zoster virus (VZV) is a human alphaherpesvirus that causes cutaneous lesions during primary and recurrent infection. Here we show that VZV disrupts PML NBs in infected cells in human skin xenografts in SCID mice and that the disruption is achieved by open reading frame 61 (ORF61) protein via its SUMO-interacting motifs (SIMs). Three conserved SIMs mediated ORF61 binding to SUMO1 and were required for ORF61 association with and disruption of PML NBs. Mutation of the ORF61 SIMs in the VZV genome showed that these motifs were necessary for PML NB dispersal in VZV-infected cells in vitro. In vivo, PML NBs were highly abundant, especially in basal layer cells of uninfected skin, whereas their frequency was significantly decreased in VZV-infected cells. In contrast, mutation of the ORF61 SIMs reduced ORF61 association with PML NBs, most PML NBs remained intact and importantly, viral replication in skin was severely impaired. The ORF61 SIM mutant virus failed to cause the typical VZV lesions that penetrate across the basement membrane into the dermis and viral spread in the epidermis was limited. These experiments indicate that VZV pathogenesis in skin depends upon the ORF61-mediated disruption of PML NBs and that the ORF61 SUMO-binding function is necessary for this effect. More broadly, our study elucidates the importance of PML NBs for the innate control of a viral pathogen during infection of differentiated cells within their tissue microenvironment in vivo and the requirement for a viral protein with SUMO-binding capacity to counteract this intrinsic barrier.


Assuntos
Herpesvirus Humano 3/genética , Herpesvirus Humano 3/patogenicidade , Corpos de Inclusão Intranuclear/metabolismo , Pele/virologia , Proteínas Virais/genética , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Clonagem Molecular , Genes Virais , Herpesvirus Humano 3/fisiologia , Humanos , Corpos de Inclusão Intranuclear/virologia , Leucemia Promielocítica Aguda , Camundongos , Camundongos SCID , Modelos Animais , Mutagênese , Plasmídeos/genética , Domínios e Motivos de Interação entre Proteínas/genética , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Regulação para Cima , Proteínas Virais/metabolismo , Replicação Viral
16.
Nat Commun ; 14(1): 7958, 2023 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-38042814

RESUMO

Herpesviruses remain a burden for animal and human health, including the medically important varicella-zoster virus (VZV). Membrane fusion mediated by conserved core glycoproteins, the fusogen gB and the heterodimer gH-gL, enables herpesvirus cell entry. The ectodomain of gB orthologs has five domains and is proposed to transition from a prefusion to postfusion conformation but the functional relevance of the domains for this transition remains poorly defined. Here we describe structure-function studies of the VZV gB DIII central helix targeting residues 526EHV528. Critically, a H527P mutation captures gB in a prefusion conformation as determined by cryo-EM, a loss of membrane fusion in a virus free assay, and failure of recombinant VZV to spread in cell monolayers. Importantly, two predominant cryo-EM structures of gB[H527P] are identified by 3D classification and focused refinement, suggesting they represented gB conformations in transition. These studies reveal gB DIII as a critical element for herpesvirus gB fusion function.


Assuntos
Herpesvirus Humano 1 , Proteínas do Envelope Viral , Animais , Humanos , Proteínas do Envelope Viral/metabolismo , Mutagênese , Mutação , Herpesvirus Humano 3/genética , Herpesvirus Humano 1/genética , Internalização do Vírus
17.
J Virol ; 85(9): 4095-110, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21345964

RESUMO

Varicella-zoster virus (VZV) is the alphaherpesvirus that causes chicken pox (varicella) and shingles (zoster). The two VZV glycoproteins gE and gI form a heterodimer that mediates efficient cell-to-cell spread. Deletion of gI yields a small-plaque-phenotype virus, ΔgI virus, which is avirulent in human skin using the xenograft model of VZV pathogenesis. In the present study, 10 mutant viruses were generated to determine which residues were required for the typical function of gI. Three phosphorylation sites in the cytoplasmic domain of gI were not required for VZV virulence in vivo. Two deletion mutants mapped a gE binding region in gI to residues 105 to 125. A glycosylation site, N116, in this region did not affect virulence. Substitution of four cysteine residues highly conserved in the Alphaherpesvirinae established that C95 is required for gE/gI heterodimer formation. The C95A and Δ105-125 (with residues 105 to 125 deleted) viruses had small-plaque phenotypes with reduced replication kinetics in vitro similar to those of the ΔgI virus. The Δ105-125 virus was avirulent for human skin in vivo. In contrast, the C95A mutant replicated in vivo but with significantly reduced kinetics compared to those of the wild-type virus. In addition to abolished gE/gI heterodimer formation, gI from the C95A or the Δ105-125 mutant was not recognized by monoclonal antibodies that detect the canonical conformation of gI, demonstrating structural disruption of gI in these viruses. This alteration prevented gI incorporation into virus particles. Thus, residues C95 and 105 to 125 are critical for gI structure required for gE/gI heterodimer formation, virion incorporation, and ultimately, effective viral spread in human skin.


Assuntos
Herpesvirus Humano 3/patogenicidade , Mapeamento de Interação de Proteínas , Multimerização Proteica , Pele/virologia , Proteínas do Envelope Viral/metabolismo , Fatores de Virulência/metabolismo , Substituição de Aminoácidos , Linhagem Celular , Cisteína/genética , Análise Mutacional de DNA , Herpesvirus Humano 3/genética , Humanos , Deleção de Sequência , Pele/patologia , Proteínas do Envelope Viral/genética , Ensaio de Placa Viral , Virulência , Fatores de Virulência/genética , Replicação Viral
18.
J Virol ; 84(1): 141-52, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19828615

RESUMO

Varicella-zoster virus (VZV) infection is usually mild in healthy individuals but can cause severe disease in immunocompromised patients. Prophylaxis with varicella-zoster immunoglobulin can reduce the severity of VZV if given shortly after exposure. Glycoprotein H (gH) is a highly conserved herpesvirus protein with functions in virus entry and cell-cell spread and is a target of neutralizing antibodies. The anti-gH monoclonal antibody (MAb) 206 neutralizes VZV in vitro. To determine the requirement for gH in VZV pathogenesis in vivo, MAb 206 was administered to SCID mice with human skin xenografts inoculated with VZV. Anti-gH antibody given at 6 h postinfection significantly reduced the frequency of skin xenograft infection by 42%. Virus titers, genome copies, and lesion size were decreased in xenografts that became infected. In contrast, administering anti-gH antibody at 4 days postinfection suppressed VZV replication but did not reduce the frequency of infection. The neutralizing anti-gH MAb 206 blocked virus entry, cell fusion, or both in skin in vivo. In vitro, MAb 206 bound to plasma membranes and to surface virus particles. Antibody was internalized into vacuoles within infected cells, associated with intracellular virus particles, and colocalized with markers for early endosomes and multivesicular bodies but not the trans-Golgi network. MAb 206 blocked spread, altered intracellular trafficking of gH, and bound to surface VZV particles, which might facilitate their uptake and targeting for degradation. As a consequence, antibody interference with gH function would likely prevent or significantly reduce VZV replication in skin during primary or recurrent infection.


Assuntos
Anticorpos Monoclonais/farmacocinética , Anticorpos Antivirais/uso terapêutico , Herpesvirus Humano 3/patogenicidade , Glicoproteínas de Membrana/imunologia , Pele/virologia , Proteínas Virais/imunologia , Animais , Anticorpos Monoclonais/uso terapêutico , Transporte Biológico , Herpesvirus Humano 3/efeitos dos fármacos , Herpesvirus Humano 3/imunologia , Humanos , Imunoglobulinas/uso terapêutico , Camundongos , Camundongos SCID , Pele/imunologia , Transplante de Pele , Transplante Heterólogo
19.
J Virol ; 83(15): 7495-506, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19474103

RESUMO

Glycoprotein B (gB), the most conserved protein in the family Herpesviridae, is essential for the fusion of viral and cellular membranes. Information about varicella-zoster virus (VZV) gB is limited, but homology modeling showed that the structure of VZV gB was similar to that of herpes simplex virus (HSV) gB, including the putative fusion loops. In contrast to HSV gB, VZV gB had a furin recognition motif ([R]-X-[KR]-R-|-X, where | indicates the position at which the polypeptide is cleaved) at residues 491 to 494, thought to be required for gB cleavage into two polypeptides. To investigate their contribution, the putative primary fusion loop or the furin recognition motif was mutated in expression constructs and in the context of the VZV genome. Substitutions in the primary loop, W180G and Y185G, plus the deletion mutation Delta491RSRR494 and point mutation 491GSGG494 in the furin recognition motif did not affect gB expression or cellular localization in transfected cells. Infectious VZV was recovered from parental Oka (pOka)-bacterial artificial chromosomes that had either the Delta491RSRR494 or 491GSGG494 mutation but not the point mutations W180G and Y185G, demonstrating that residues in the primary loop of gB were essential but gB cleavage was not required for VZV replication in vitro. Virion morphology, protein localization, plaque size, and replication were unaffected for the pOka-gBDelta491RSRR494 or pOka-gB491GSGG494 virus compared to pOka in vitro. However, deletion of the furin recognition motif caused attenuation of VZV replication in human skin xenografts in vivo. This is the first evidence that cleavage of a herpesvirus fusion protein contributes to viral pathogenesis in vivo, as seen for fusion proteins in other virus families.


Assuntos
Varicela/virologia , Furina/metabolismo , Herpesvirus Humano 3/patogenicidade , Mutagênese , Pele/virologia , Proteínas do Envelope Viral/genética , Replicação Viral , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Varicela/metabolismo , Varicela/patologia , Herpesvirus Humano 3/química , Herpesvirus Humano 3/genética , Herpesvirus Humano 3/fisiologia , Humanos , Técnicas In Vitro , Camundongos , Camundongos SCID , Dados de Sequência Molecular , Mutação , Ligação Proteica , Alinhamento de Sequência , Pele/metabolismo , Pele/patologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo
20.
J Med Virol ; 82(7): 1241-6, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20513091

RESUMO

Serum antibodies to bovine norovirus have been found recently in about 22% of humans. Whether this prevalence reflects limited virulence properties of the virus or that inherited host factors provide protection against bovine norovirus infection in humans remains to be established. To investigate whether histo-blood group antigens correlate with the presence of bovine norovirus (GIII.2) antibody, plasma (n = 105) from Swedish blood donors, genotyped and phenotyped for secretor, Lewis and ABO, were tested and compared for the frequency of IgG antibody and antibody titer to Bo/Newbury2/76/UK. In total, 26.7% (28/105) of Swedish blood donors were antibody-positive. Two non-secretors (2/21, 9.5%) were antibody-positive compared with 26/84 (31%) secretors (P = 0.047). While no statistically significant correlation was found between the frequency of antibodies to bovine norovirus and different ABO blood groups, individuals with blood type B presented the highest frequency of antibodies (37.5%) compared with 0-30% among other blood groups. Individuals with Le(a-b+) had not only higher frequency of antibodies (31.3%) compared with Le(a+b-) (11%) (P = 0.068) but also higher antibody titer (P = 0.085) although this was not significant statistically. No detectable cross-reaction between bovine GIII.2 and human GII.3 NoV VLP was found with human and animal sera. The results of this study suggest that bovine norovirus infections occur in Sweden and that secretor status but not ABO blood groups is a possible risk factor for infection.


Assuntos
Sistema ABO de Grupos Sanguíneos , Anticorpos Antivirais/sangue , Infecções por Caliciviridae/sangue , Infecções por Caliciviridae/epidemiologia , Norovirus/imunologia , Adulto , Idoso , Animais , Anticorpos Antivirais/imunologia , Doadores de Sangue , Infecções por Caliciviridae/genética , Bovinos , Reações Cruzadas , Fucosiltransferases/genética , Genótipo , Humanos , Pessoa de Meia-Idade , Fatores de Risco , Suécia/epidemiologia , Galactosídeo 2-alfa-L-Fucosiltransferase
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa