Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 228
Filtrar
1.
Cells ; 10(11)2021 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-34831342

RESUMO

The regulation of the nucleocytoplasmic release of herpesviral capsids is defined by the process of nuclear egress. Due to their large size, nuclear capsids are unable to traverse via nuclear pores, so that herpesviruses evolved to develop a vesicular transport pathway mediating their transition through both leaflets of the nuclear membrane. This process involves regulatory proteins, which support the local distortion of the nuclear envelope. For human cytomegalovirus (HCMV), the nuclear egress complex (NEC) is determined by the pUL50-pUL53 core that initiates multicomponent assembly with NEC-associated proteins and capsids. Hereby, pUL50 serves as a multi-interacting determinant that recruits several viral and cellular factors by direct and indirect contacts. Recently, we generated an ORF-UL50-deleted recombinant HCMV in pUL50-complementing cells and obtained first indications of putative additional functions of pUL50. In this study, we produced purified ΔUL50 particles under both complementing (ΔUL50C) and non-complementing (ΔUL50N) conditions and performed a phenotypical characterization. Findings were as follows: (i) ΔUL50N particle preparations exhibited a clear replicative defect in qPCR-based infection kinetics compared to ΔUL50C particles; (ii) immuno-EM analysis of ΔUL50C did not reveal major changes in nuclear distribution of pUL53 and lamin A/C; (iii) mass spectrometry-based quantitative proteomics showed a large concordance of protein contents in the NIEP fractions of ΔUL50C and ΔUL50N particles, but virion fraction was close to the detection limit for ΔUL50N; (iv) confocal imaging of viral marker proteins of immediate early (IE) and later phases of ΔUL50N infection indicated a very low number of cells showing an onset of viral lytic protein expression; and, finally (v) quantitative measurements of encapsidated genomes provided evidence for a substantial reduction in the DNA contents in ΔUL50N compared to ΔUL50C particles. In summary, the results point to a complex and important regulatory role of the HCMV nuclear egress protein pUL50 in the maturation of infectious virus.


Assuntos
Núcleo Celular/metabolismo , Citomegalovirus/patogenicidade , Proteínas Virais/metabolismo , Capsídeo/metabolismo , Capsídeo/ultraestrutura , Linhagem Celular , Citomegalovirus/genética , Citomegalovirus/ultraestrutura , Empacotamento do DNA/genética , Fibroblastos/metabolismo , Fibroblastos/virologia , Regulação Viral da Expressão Gênica , Genes Precoces , Genoma Viral , Humanos , Cinética , Membrana Nuclear/metabolismo , Proteômica , Proteínas Virais/ultraestrutura , Vírion/metabolismo , Vírion/ultraestrutura , Replicação Viral/fisiologia
2.
Nat Commun ; 12(1): 5513, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34535641

RESUMO

Under the Baltimore nucleic acid-based virus classification scheme, the herpesvirus human cytomegalovirus (HCMV) is a Class I virus, meaning that it contains a double-stranded DNA genome-and no RNA. Here, we report sub-particle cryoEM reconstructions of HCMV virions at 2.9 Å resolution revealing structures resembling non-coding transfer RNAs (tRNAs) associated with the virion's capsid-bound tegument protein, pp150. Through deep sequencing, we show that these RNA sequences match human tRNAs, and we built atomic models using the most abundant tRNA species. Based on our models, tRNA recruitment is mediated by the electrostatic interactions between tRNA phosphate groups and the helix-loop-helix motif of HCMV pp150. The specificity of these interactions may explain the absence of such tRNA densities in murine cytomegalovirus and other human herpesviruses.


Assuntos
Capsídeo/metabolismo , Citomegalovirus/ultraestrutura , Fosfoproteínas/metabolismo , RNA de Transferência/metabolismo , Proteínas da Matriz Viral/metabolismo , Vírion/ultraestrutura , Anticódon/metabolismo , Sequência de Bases , Linhagem Celular , Microscopia Crioeletrônica , Glutamato-tRNA Ligase/química , Glutamato-tRNA Ligase/metabolismo , Humanos , Modelos Moleculares , Fosfoproteínas/ultraestrutura , RNA Viral/ultraestrutura , Proteínas da Matriz Viral/ultraestrutura
3.
Nat Commun ; 12(1): 4538, 2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34315863

RESUMO

How the human cytomegalovirus (HCMV) genome-the largest among human herpesviruses-is packaged, retained, and ejected remains unclear. We present the in situ structures of the symmetry-mismatched portal and the capsid vertex-specific components (CVSCs) of HCMV. The 5-fold symmetric 10-helix anchor-uncommon among known portals-contacts the portal-encircling DNA, which is presumed to squeeze the portal as the genome packaging proceeds. We surmise that the 10-helix anchor dampens this action to delay the portal reaching a "head-full" packaging state, thus facilitating the large genome to be packaged. The 6-fold symmetric turret, latched via a coiled coil to a helix from a major capsid protein, supports the portal to retain the packaged genome. CVSCs at the penton vertices-presumed to increase inner capsid pressure-display a low stoichiometry, which would aid genome retention. We also demonstrate that the portal and capsid undergo conformational changes to facilitate genome ejection after viral cell entry.


Assuntos
Citomegalovirus/química , Citomegalovirus/genética , Empacotamento do DNA/genética , Genoma Viral , Capsídeo/química , Capsídeo/ultraestrutura , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Citomegalovirus/ultraestrutura , DNA Viral/genética , DNA Viral/ultraestrutura , Humanos , Modelos Moleculares , Homologia Estrutural de Proteína , Vírion/química , Vírion/ultraestrutura
4.
Cell ; 184(14): 3774-3793.e25, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34115982

RESUMO

Cytomegaloviruses (CMVs) have co-evolved with their mammalian hosts for millions of years, leading to remarkable host specificity and high infection prevalence. Macrophages, which already populate barrier tissues in the embryo, are the predominant immune cells at potential CMV entry sites. Here we show that, upon CMV infection, macrophages undergo a morphological, immunophenotypic, and metabolic transformation process with features of stemness, altered migration, enhanced invasiveness, and provision of the cell cycle machinery for viral proliferation. This complex process depends on Wnt signaling and the transcription factor ZEB1. In pulmonary infection, mouse CMV primarily targets and reprograms alveolar macrophages, which alters lung physiology and facilitates primary CMV and secondary bacterial infection by attenuating the inflammatory response. Thus, CMV profoundly perturbs macrophage identity beyond established limits of plasticity and rewires specific differentiation processes, allowing viral spread and impairing innate tissue immunity.


Assuntos
Citomegalovirus/fisiologia , Macrófagos Alveolares/virologia , Animais , Apresentação de Antígeno , Efeito Espectador , Ciclo Celular , Linhagem Celular Transformada , Reprogramação Celular , Citomegalovirus/patogenicidade , Citomegalovirus/ultraestrutura , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Proteínas de Fluorescência Verde/metabolismo , Pulmão/patologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/ultraestrutura , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fenótipo , Células-Tronco/patologia , Replicação Viral/fisiologia , Via de Sinalização Wnt
5.
J Cell Physiol ; 236(9): 6726-6741, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33611789

RESUMO

Thrombocytopenia is a common complication of human cytomegalovirus (HCMV) infection in immunocompromised hosts, which contributes to poor prognosis even in patients receiving antiviral treatment. Here, we investigated the megakaryo/thrombopoiesis process, including the involvement of the c-Mpl/IEX-1 pathway, after HCMV infection, identified receptors mediating the interaction between megakaryocytes (MKs) and HCMV, and explored novel therapeutic targets. Our data shows that HCMV directly infects megakaryocytes in patients with HCMV DNAemia and influences megakaryopoiesis via the c-Mpl/IEX-1 pathway throughout megakaryocyte maturation, apoptosis, and platelet generation in vivo and in vitro. After treatment with inhibitors of PDGFRα and αvß3, the HCMV infection rate in MKs was significantly reduced, suggesting that IMC-3G3 and anti-αvß3 are potential therapeutic alternatives for viral infection. In summary, our study proposes a possible mechanism and potential treatments for thrombocytopenia caused by HCMV infection and other viral diseases associated with abnormal hemostasis.


Assuntos
Citomegalovirus/fisiologia , Transplante de Células-Tronco Hematopoéticas , Integrina alfaVbeta3/metabolismo , Megacariócitos/virologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores de Trombopoetina/metabolismo , Transdução de Sinais , Trombopoese , Adolescente , Adulto , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Criança , Citomegalovirus/ultraestrutura , Infecções por Citomegalovirus/patologia , Regulação para Baixo , Feminino , Humanos , Masculino , Proteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Análise Multivariada , Ploidias , Fatores de Risco , Receptor 2 Toll-Like/metabolismo , Transplante Homólogo , Adulto Jovem
6.
Cell Microbiol ; 23(2): e13280, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33073426

RESUMO

Detailed analysis of secondary envelopment of the herpesvirus human cytomegalovirus (HCMV) by transmission electron microscopy (TEM) is crucial for understanding the formation of infectious virions. Here, we present a convolutional neural network (CNN) that automatically recognises cytoplasmic capsids and distinguishes between three HCMV capsid envelopment stages in TEM images. 315 TEM images containing 2,610 expert-labelled capsids of the three classes were available for CNN training. To overcome the limitation of small training datasets and thus poor CNN performance, we used a deep learning method, the generative adversarial network (GAN), to automatically increase our labelled training dataset with 500 synthetic images and thus to 9,192 labelled capsids. The synthetic TEM images were added to the ground truth dataset to train the Faster R-CNN deep learning-based object detector. Training with 315 ground truth images yielded an average precision (AP) of 53.81% for detection, whereas the addition of 500 synthetic training images increased the AP to 76.48%. This shows that generation and additional use of synthetic labelled images for detector training is an inexpensive way to improve detector performance. This work combines the gold standard of secondary envelopment research with state-of-the-art deep learning technology to speed up automatic image analysis even when large labelled training datasets are not available.


Assuntos
Capsídeo/ultraestrutura , Citomegalovirus/ultraestrutura , Aprendizado Profundo , Infecções por Herpesviridae/diagnóstico por imagem , Processamento de Imagem Assistida por Computador/métodos , Vírion/ultraestrutura , Algoritmos , Citomegalovirus/metabolismo , Infecções por Herpesviridae/virologia , Humanos , Aprendizado de Máquina , Microscopia Eletrônica de Transmissão , Redes Neurais de Computação , Vírion/metabolismo
7.
Virology ; 540: 57-65, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31739185

RESUMO

Among the Herpesviridae, human cytomegalovirus (HCMV) owns the largest genome and displays a huge coding potential. Here, we characterized the UL5 gene product (pUL5) of the clinical isolate TR strain. The protein was predicted as a 166-amino-acid membrane protein with a theoretical mass of 19 kDa. Recombinant virus expressing pUL5 with a tag allowed the identification of two pUL5 non-glycosylated species of approximately 19 and 9 kDa, expressed with early and late kinetic respectively. Experiments in infection confirmed that the lower molecular weight species was translated from an internal ATG in the UL5 open reading frame. Confocal microscopy analysis showed that pUL5 localized within the assembly compartment, but is not incorporated in the virion, as shown by Western blot on purified viral particles. Finally, pull-down experiments coupled with mass spectrometry analysis identified IQGAP1 as a pUL5 interactor, giving new hints on possible roles of pUL5 during HCMV infection.


Assuntos
Infecções por Citomegalovirus/metabolismo , Infecções por Citomegalovirus/virologia , Citomegalovirus/fisiologia , Interações Hospedeiro-Patógeno , Proteínas Virais/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Células Cultivadas , Citomegalovirus/isolamento & purificação , Citomegalovirus/ultraestrutura , Regulação Viral da Expressão Gênica , Humanos , Fases de Leitura Aberta , Ligação Proteica , Transporte Proteico , RNA Viral , Transcrição Gênica
8.
J Virol ; 93(13)2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30996102

RESUMO

Human cytomegalovirus (HCMV) secondary envelopment requires the viral tegument protein pUL71. The lack of pUL71 results in a complex ultrastructural phenotype with increased numbers of viral capsids undergoing envelopment at the cytoplasmic virus assembly complex. Here, we report a role of the pUL71 C terminus in secondary envelopment. Mutant viruses expressing C-terminally truncated pUL71 (TB71del327-361 and TB71del348-351) exhibited an impaired secondary envelopment in transmission electron microscopy (TEM) studies. Further mutational analyses of the C terminus revealed a tetralysine motif whose mutation (TB71mutK348-351A) resulted in an envelopment defect that was undistinguishable from the defect caused by truncation of the pUL71 C terminus. Interestingly, not all morphological alterations that define the ultrastructural phenotype of a TB71stop virus were found in cells infected with the C-terminally mutated viruses. This suggests that pUL71 provides additional functions that modulate HCMV morphogenesis and are harbored elsewhere in pUL71. This is also reflected by an intermediate growth defect of the C-terminally mutated viruses compared to the growth of the TB71stop virus. Electron tomography and three-dimensional visualization of different stages of secondary envelopment in TB71mutK348-351A-infected cells showed unambiguously the formation of a bud neck. Furthermore, we provide evidence for progressive tegument formation linked to advancing grades of capsid envelopment, suggesting that tegumentation and envelopment are intertwined processes. Altogether, we identified the importance of the pUL71 C terminus and, specifically, of a positively charged tetralysine motif for HCMV secondary envelopment.IMPORTANCE Human cytomegalovirus (HCMV) is an important human pathogen that causes severe symptoms, especially in immunocompromised hosts. Furthermore, congenital HCMV infection is the leading viral cause of severe birth defects. Development of antiviral drugs to prevent the production of infectious virus progeny is challenging due to a complex and multistep virion morphogenesis. The mechanism of secondary envelopment is still not fully understood; nevertheless, it represents a potential target for antiviral drugs. Our identification of the role of a positively charged motif in the pUL71 C terminus for efficient HCMV secondary envelopment underlines the importance of pUL71 and, especially, its C terminus for this process. It furthermore shows how cell-associated spread and virion release depend on secondary envelopment. Ultrastructural analyses of different stages of envelopment contribute to a better understanding of the mechanisms underlying the process of secondary envelopment. This may bring us closer to the development of novel concepts to treat HCMV infections.


Assuntos
Infecções por Citomegalovirus/metabolismo , Citomegalovirus/genética , Citomegalovirus/fisiologia , Polilisina , Capsídeo/metabolismo , Capsídeo/ultraestrutura , Proteínas do Capsídeo/ultraestrutura , Linhagem Celular , Citomegalovirus/ultraestrutura , Citoplasma/virologia , Humanos , Mutação , Alinhamento de Sequência , Proteínas Virais/metabolismo , Montagem de Vírus
9.
PLoS Pathog ; 14(12): e1007452, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30507948

RESUMO

Human cytomegalovirus (HCMV) enters host by glycoprotein B (gB)-mediated membrane fusion upon receptor-binding to gH/gL-related complexes, causing devastating diseases such as birth defects. Although an X-ray crystal structure of the recombinant gB ectodomain at postfusion conformation is available, the structures of prefusion gB and its complex with gH/gL on the viral envelope remain elusive. Here, we demonstrate the utility of cryo electron tomography (cryoET) with energy filtering and the cutting-edge technologies of Volta phase plate (VPP) and direct electron-counting detection to capture metastable prefusion viral fusion proteins and report the structures of glycoproteins in the native environment of HCMV virions. We established the validity of our approach by obtaining cryoET in situ structures of the vesicular stomatitis virus (VSV) glycoprotein G trimer (171 kD) in prefusion and postfusion conformations, which agree with the known crystal structures of purified G trimers in both conformations. The excellent contrast afforded by these technologies has enabled us to identify gB trimers (303kD) in two distinct conformations in HCMV tomograms and obtain their in situ structures at up to 21 Å resolution through subtomographic averaging. The predominant conformation (79%), which we designate as gB prefusion conformation, fashions a globular endodomain and a Christmas tree-shaped ectodomain, while the minority conformation (21%) has a columnar tree-shaped ectodomain that matches the crystal structure of the "postfusion" gB ectodomain. We also observed prefusion gB in complex with an "L"-shaped density attributed to the gH/gL complex. Integration of these structures of HCMV glycoproteins in multiple functional states and oligomeric forms with existing biochemical data and domain organization of other class III viral fusion proteins suggests that gH/gL receptor-binding triggers conformational changes of gB endodomain, which in turn triggers two essential steps to actuate virus-cell membrane fusion: exposure of gB fusion loops and unfurling of gB ectodomain.


Assuntos
Citomegalovirus/fisiologia , Tomografia com Microscopia Eletrônica/métodos , Proteínas do Envelope Viral/ultraestrutura , Internalização do Vírus , Citomegalovirus/química , Citomegalovirus/ultraestrutura , Infecções por Citomegalovirus/transmissão , Humanos , Conformação Proteica
10.
J Struct Biol ; 204(3): 406-419, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30352275

RESUMO

Human cytomegalovirus (HCMV) entry into susceptible cells is a fast intricate process that is not fully understood. Although, previous studies explored different aspects of this process by means of biochemical and inhibitors assays, a clear morphological characterization of its steps at the ultrastructural level is still lacking. We attempted to characterize those intermediates involved during HCMV entry by developing a methodological approach that resulted in optimal ultrastructure preservation and allowed for 3D imaging. It involves rapid freezing and cryosubstitution which ensure a clear visibility of membranous leaflets as well as retained membranous continuity. Likewise, it delivered a reproducible optimization of the growth and infection conditions that are pivotal towards maintaining biologically active enriched input virus particles. Data acquisition was achieved through STEM tomography in a 3D context. Indeed, several intermediates that characterize HCMV entry-related events were observed both extra- and intracellularly. Some of the cell-membrane associated viral particles that we referred to as "Pinocchio particles" were morphologically altered in comparison to the cell-free virions. We were also able to characterize intracellular fusion intermediates taking place between the viral envelope and the vesicular membranes. Furthermore, inhibiting actin polymerization by Latrunculin-A enabled us to spot fusion-like intermediates of the viral envelope with the host cell plasma membrane that we did not observe in the untreated infected cells. Our data also suggests that Dyngo-4a; a dynamin-2 inhibitor, does not interfere with the internalization of the HCMV into the host cells as previously deduced.


Assuntos
Citomegalovirus/fisiologia , Tomografia com Microscopia Eletrônica/métodos , Vírion/fisiologia , Internalização do Vírus , Linhagem Celular , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Membrana Celular/virologia , Células Cultivadas , Citomegalovirus/ultraestrutura , Dinamina II/antagonistas & inibidores , Dinamina II/metabolismo , Fibroblastos/metabolismo , Fibroblastos/ultraestrutura , Fibroblastos/virologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Hidrazonas/farmacologia , Imageamento Tridimensional/métodos , Naftóis/farmacologia , Reprodutibilidade dos Testes , Vírion/ultraestrutura
11.
Viruses ; 10(1)2018 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-29342872

RESUMO

The nuclear phase of herpesvirus replication is regulated through the formation of regulatory multi-component protein complexes. Viral genomic replication is followed by nuclear capsid assembly, DNA encapsidation and nuclear egress. The latter has been studied intensely pointing to the formation of a viral core nuclear egress complex (NEC) that recruits a multimeric assembly of viral and cellular factors for the reorganization of the nuclear envelope. To date, the mechanism of the association of human cytomegalovirus (HCMV) capsids with the NEC, which in turn initiates the specific steps of nuclear capsid budding, remains undefined. Here, we provide electron microscopy-based data demonstrating the association of both nuclear capsids and NEC proteins at nuclear lamina budding sites. Specifically, immunogold labelling of the core NEC constituent pUL53 and NEC-associated viral kinase pUL97 suggested an intranuclear NEC-capsid interaction. Staining patterns with phospho-specific lamin A/C antibodies are compatible with earlier postulates of targeted capsid egress at lamina-depleted areas. Important data were provided by co-immunoprecipitation and in vitro kinase analyses using lysates from HCMV-infected cells, nuclear fractions, or infectious virions. Data strongly suggest that nuclear capsids interact with pUL53 and pUL97. Combined, the findings support a refined concept of HCMV nuclear trafficking and NEC-capsid interaction.


Assuntos
Capsídeo/fisiologia , Citomegalovirus/enzimologia , Citomegalovirus/fisiologia , Membrana Nuclear/virologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Quinases/metabolismo , Núcleo Celular/ultraestrutura , Núcleo Celular/virologia , Citomegalovirus/ultraestrutura , Interações Hospedeiro-Patógeno , Humanos , Imuno-Histoquímica , Microscopia Eletrônica , Membrana Nuclear/ultraestrutura , Lâmina Nuclear/ultraestrutura , Lâmina Nuclear/virologia , Fosforilação , Proteínas Virais/metabolismo , Montagem de Vírus , Liberação de Vírus , Replicação Viral
12.
Virology ; 514: 165-169, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29190455

RESUMO

Glossina palipides salivary gland hypertrophy virus (GpSGHV) infects tsetse flies, which are vectors for African trypanosomosis. This virus represents a major challenge in insect mass rearing and has hampered the implementation of the sterile insect technique programs in the Member States of the International Atomic Energy Agency. GpSGHV virions consist of long rod-shaped particles over 9000Å in length, but little is known about their detailed structural organization. We show by cryo electron microscopy and cryo electron tomography that the GpSGHV virion has a unique, non-icosahedral helical structure. Its envelope exhibits regularly spaced spikes that protrude from the lipid bilayer and are arranged on a four-start helix. This study provides a detailed insight into the 3D architecture of GpSGHV, which will help to understand the viral life cycle and possibly allow the design of antiviral strategies in the context of tsetse fly infections.


Assuntos
Citomegalovirus/ultraestrutura , Moscas Tsé-Tsé/virologia , Animais , Microscopia Crioeletrônica , Citomegalovirus/fisiologia , Vírus de Insetos/fisiologia , Vírus de Insetos/ultraestrutura , Masculino , Vírion/fisiologia , Vírion/ultraestrutura
13.
PLoS One ; 12(1): e0169580, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28060895

RESUMO

Widely used methods for quantification of human cytomegalovirus (HCMV) infection in cell culture such as immunoblotting or plaque reduction assays are generally restricted to low throughput and require time-consuming evaluation. Up to now, only few HCMV reporter cell lines have been generated to overcome these restrictions and they are afflicted with other limitations because permanently expandable cell lines are normally not fully permissive to HCMV. In this work, a previously existing epithelial cell line hosting a luciferase gene under control of a Varicella-zoster virus promoter was adopted to investigate HCMV infection. The cells were susceptible to different HCMV strains at infection efficiencies that corresponded to their respective degree of epithelial cell tropism. Expression of early and late viral antigens, formation of nuclear inclusions, release of infectious virus progeny, and focal growth indicated productive viral replication. However, viral release and spread occurred at lower levels than in primary cell lines which appears to be due to a malfunction of virion morphogenesis during the nuclear stage. Expression of the luciferase reporter gene was specifically induced in HCMV infected cultures as a function of the virus dose and dependent on viral immediate early gene expression. The level of reporter activity accurately reflected infection efficiencies as determined by viral antigen immunostaining, and hence could discriminate the cell tropism of the tested virus strains. As proof-of-principle, we demonstrate that this cell line is applicable to evaluate drug resistance of clinical HCMV isolates and the neutralization capacity of human sera, and that it allows comparative and simultaneous analysis of HCMV and human herpes simplex virus type 1. In summary, the permanent epithelial reporter cell line allows robust, rapid and objective quantitation of HCMV infection and it will be particularly useful in higher throughput analyses as well as in comparative analyses of different human herpesviruses.


Assuntos
Alphaherpesvirinae/genética , Citomegalovirus/fisiologia , Expressão Gênica , Genes Reporter , Proteínas Imediatamente Precoces/metabolismo , Luciferases/genética , Regiões Promotoras Genéticas , Animais , Antígenos Virais/imunologia , Antígenos Virais/metabolismo , Linhagem Celular , Citomegalovirus/ultraestrutura , Genoma Viral , Humanos , Proteínas Imediatamente Precoces/imunologia , Tropismo Viral , Vírion/ultraestrutura , Replicação Viral
14.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27795411

RESUMO

The glycoprotein O (gO) is betaherpesvirus specific. Together with the viral glycoproteins H and L, gO forms a covalent trimeric complex that is part of the viral envelope. This trimer is crucial for cell-free infectivity of human cytomegalovirus (HCMV) but dispensable for cell-associated spread. We hypothesized that the amino acids that are conserved among gOs of different cytomegaloviruses are important for the formation of the trimeric complex and hence for efficient virus spread. In a mutational approach, nine peptide sites, containing all 13 highly conserved amino acids, were analyzed in the context of HCMV strain TB40-BAC4 with regard to infection efficiency and formation of the gH/gL/gO complex. Mutation of amino acids (aa) 181 to 186 or aa 193 to 198 resulted in the loss of the trimer and a complete small-plaque phenotype, whereas mutation of aa 108 or aa 249 to 254 caused an intermediate phenotype. While individual mutations of the five conserved cysteines had little impact, their relevance was revealed in a combined mutation, which abrogated both complex formation and cell-free infectivity. C343 was unique, as it was sufficient and necessary for covalent binding of gO to gH/gL. Remarkably, however, C218 together with C167 rescued infectivity in the absence of detectable covalent complex formation. We conclude that all highly conserved amino acids contribute to the function of gO to some extent but that aa 181 to 198 and cysteines 343, 218, and 167 are particularly relevant. Surprisingly, covalent binding of gO to gH/gL is required neither for its incorporation into virions nor for proper function in cell-free infection. IMPORTANCE: Like all herpesviruses, the widespread human pathogen HCMV depends on glycoproteins gB, gH, and gL for entry into target cells. Additionally, gH and gL have to bind gO in a trimeric complex for efficient cell-free infection. Homologs of gO are shared by all cytomegaloviruses, with 13 amino acids being highly conserved. In a mutational approach we analyzed these amino acids to elucidate their role in the function of gO. All conserved amino acids contributed either to formation of the trimeric complex or to cell-free infection. Notably, these two phenotypes were not inevitably linked as the mutation of a charged cluster in the center of gO abrogated cell-free infection while trimeric complexes were still being formed. Cysteine 343 was essential for covalent binding of gO to gH/gL; however, noncovalent complex formation in the absence of cysteine 343 also allowed for cell-free infectivity.


Assuntos
Aminoácidos/química , Citomegalovirus/química , Glicoproteínas de Membrana/química , Proteínas do Envelope Viral/química , Vírion/química , Sequência de Aminoácidos , Aminoácidos/metabolismo , Animais , Linhagem Celular , Clonagem Molecular , Sequência Conservada , Citomegalovirus/metabolismo , Citomegalovirus/ultraestrutura , Células Endoteliais/ultraestrutura , Células Endoteliais/virologia , Escherichia coli/genética , Escherichia coli/metabolismo , Fibroblastos/ultraestrutura , Fibroblastos/virologia , Expressão Gênica , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Mutação , Cultura Primária de Células , Multimerização Proteica , Proteínas Recombinantes , Alinhamento de Sequência , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Vírion/metabolismo , Vírion/ultraestrutura
15.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27795424

RESUMO

Formation of the cytoplasmic viral assembly compartment (cVAC) is an important step for efficient human cytomegalovirus (HCMV) assembly. To do this, the virus must alter and repurpose the normal cellular balance of membrane and protein flux, a process that is not well understood. Although a recent screen identified three viral proteins essential for cVAC formation, less is known about the contribution of cellular factors. We show that HCMV infection increases the protein level of a cellular trafficking factor, syntaxin 5 (STX5), a member of the syntaxin family of SNARE proteins. STX5 is recruited to the cVAC in infected cells and is required for the efficient production of infectious virions. We find that STX5 is important for normal cVAC morphology and the proper localization of viral proteins. A previously identified inhibitor of trafficking, Retro94, causes the mislocalization of STX5, an altered cVAC morphology, and dispersal of viral proteins. The presence of Retro94 results in severely impaired production of infectious virions, with a decrease as great as 5 logs. We show that this inhibition is conserved among different strains of HCMV and the various cell types that support infection, as well as for murine CMV. Thus, our data identify a key cellular trafficking factor important for supporting HCMV infection. IMPORTANCE: Human cytomegalovirus (HCMV) infection causes severe disease and mortality in immunocompromised individuals, including organ transplant and AIDS patients. In addition, infection of a developing fetus may result in lifelong complications such as deafness and learning disabilities. Understanding in detail the processes involved in HCMV replication is important for developing novel treatments. One of these essential processes, assembly of infectious virions, takes places in the cytoplasmic viral assembly compartment. We identify a cellular protein, syntaxin 5, important for generating this compartment, and show that it is required for the efficient production of infectious virions. We also show that a small molecule that disrupts this protein also significantly reduces the amount of infectious virions that are generated. Thus, by pinpointing a cellular protein that is important in the replication cycle of HCMV, we identified a novel target that can be pursued for therapeutic intervention.


Assuntos
Citomegalovirus/efeitos dos fármacos , Citoplasma/efeitos dos fármacos , Interações Hospedeiro-Patógeno , Proteínas Qa-SNARE/genética , Quinazolinas/farmacologia , Vírion/efeitos dos fármacos , Animais , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Membrana Celular/virologia , Citomegalovirus/metabolismo , Citomegalovirus/ultraestrutura , Citoplasma/metabolismo , Citoplasma/ultraestrutura , Citoplasma/virologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Células Epiteliais/virologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/ultraestrutura , Fibroblastos/virologia , Regulação da Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Transporte Proteico/efeitos dos fármacos , Proteínas Qa-SNARE/metabolismo , Transdução de Sinais , Vírion/metabolismo , Vírion/ultraestrutura , Montagem de Vírus/efeitos dos fármacos , Montagem de Vírus/genética , Proteína Vermelha Fluorescente
16.
Viruses ; 7(11): 5686-704, 2015 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-26556360

RESUMO

We show that focused ion beam/scanning electron microscopy (FIB/SEM) tomography is an excellent method to analyze the three-dimensional structure of a fibroblast nucleus infected with human cytomegalovirus (HCMV). We found that the previously described infoldings of the inner nuclear membrane, which are unique among its kind, form an extremely complex network of membrane structures not predictable by previous two-dimensional studies. In all cases they contained further invaginations (2nd and 3rd order infoldings). Quantification revealed 5498HCMV capsids within two nuclear segments, allowing an estimate of 15,000 to 30,000 capsids in the entire nucleus five days post infection. Only 0.8% proved to be enveloped capsids which were exclusively detected in 1st order infoldings (perinuclear space). Distribution of the capsids between 1st, 2nd and 3rd order infoldings is in complete agreement with the envelopment/de-envelopment model for egress of HCMV capsids from the nucleus and we confirm that capsid budding does occur at the large infoldings. Based on our results we propose the pushing membrane model: HCMV infection induces local disruption of the nuclear lamina and synthesis of new membrane material which is pushed into the nucleoplasm, forming complex membrane infoldings in a highly abundant manner, which then may be also used by nucleocapsids for budding.


Assuntos
Citomegalovirus/crescimento & desenvolvimento , Citomegalovirus/ultraestrutura , Tomografia com Microscopia Eletrônica , Membrana Nuclear/ultraestrutura , Membrana Nuclear/virologia , Tomografia Computadorizada por Raios X , Células Cultivadas , Fibroblastos/ultraestrutura , Fibroblastos/virologia , Humanos
17.
J Gen Virol ; 96(Pt 2): 239-252, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25359764

RESUMO

In human cytomegalovirus (HCMV)-infected cells, a dramatic remodelling of the nuclear architecture is linked to the creation, utilization and manipulation of subnuclear structures. This review outlines the involvement of several viral and cellular subnuclear structures in areas of HCMV replication and virus-host interaction that include viral transcription, viral DNA synthesis and the production of DNA-filled viral capsids. The structures discussed include those that promote or impede HCMV replication (such as viral replication compartments and promyelocytic leukaemia nuclear bodies, respectively) and those whose role in the infected cell is unclear (for example, nucleoli and nuclear speckles). Viral and cellular proteins associated with subnuclear structures are also discussed. The data reviewed here highlight advances in our understanding of HCMV biology and emphasize the complexity of HCMV replication and virus-host interactions in the nucleus.


Assuntos
Núcleo Celular/ultraestrutura , Núcleo Celular/virologia , Citomegalovirus/fisiologia , Citomegalovirus/ultraestrutura , Interações Hospedeiro-Patógeno , Substâncias Macromoleculares/ultraestrutura , Humanos
18.
Fertil Steril ; 101(2): 350-8, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24314922

RESUMO

OBJECTIVE: To assess the accuracy of serology to predict the presence of cytomegalovirus (CMV) in semen of homosexual men without and with HIV coinfection. DESIGN: Semen CMV was detected by electron microscopy and by polymerase chain reaction (PCR) amplification; paired serum was tested for CMV IgG/IgM. Semen HIV was detected by reverse transcription-PCR. SETTING: Licensed clinical and research laboratory. PATIENT(S): Sixty-eight men. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Frequency of CMV and HIV in semen. RESULT(S): Cytomegalovirus was detected by electron microscopy in 3 of 10 specimens examined. Forty-six (89%) of 52 HIV-infected men were seropositive for CMV by combined assay for IgG/IgM; two more (48 of 52, 92%) were seropositive for CMV IgG by separate assay; 25 (48%) of the HIV-infected men had PCR-detectable CMV DNA in at least one semen specimen, 22 of whom (42%) had CMV in all specimens. Nineteen (13%) of the 150 specimens tested positive for HIV, whereas 67 (45%) tested positive for CMV; seven specimens tested positive for both CMV and HIV. Cytomegalovirus, but not HIV, detection in semen correlated with decreased CD4(+) lymphocytes in peripheral blood (<700/µL) but was not accurately predicted by serology, leukocytospermia, or age. CONCLUSION(S): Cytomegalovirus in semen is not accurately predicted by serology. Sperm banking needs to include direct assessment of CMV in semen specimens. Strategies to eliminate CMV from semen specimens are needed to alleviate the risk of virus transmission.


Assuntos
Citomegalovirus/isolamento & purificação , HIV-1/isolamento & purificação , Homossexualidade Masculina , Sêmen/virologia , Bancos de Esperma , Adulto , Estudos de Coortes , Citomegalovirus/ultraestrutura , Infecções por HIV/sangue , Infecções por HIV/diagnóstico , Soropositividade para HIV/sangue , Soropositividade para HIV/diagnóstico , HIV-1/ultraestrutura , Humanos , Masculino , Pessoa de Meia-Idade , Bancos de Esperma/normas , Adulto Jovem
19.
Cell Microbiol ; 15(2): 305-14, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23217081

RESUMO

Electron microscopy (EM) allows visualization of viruses in fixed cells with high resolution. High-pressure freezing for sample fixation in combination with freeze substitution and embedding in resin improves significantly the preservation of cellular structures and specifically of membranes. This advancement allows better visualization of human cytomegalovirus (HCMV) morphogenesis occurring at membranes. To obtain comprehensive information on viral phenotypes from ultrastructural images it is important to also quantify morphological phenotypes. This again can be much refined by three-dimensional visualization after serial sectioning. For elucidation of dynamic processes three-dimensional tomography is extremely helpful. We analysed interaction of HCMV particles with host cell membranes during final envelopment. Both wild-type virus and a viral mutant with impaired envelopment were analysed in fibroblasts, but also using in vivo relevant human endothelial cells and macrophages. The quantification of the EM data showed similar ultrastructural phenotypes regarding the envelopment efficiency in the different cell types indicating similar mechanisms in late stages of virus morphogenesis. Furthermore, thorough analysis of the viral assembly complex (AC) - a virus-induced cytosolic structure - by using three-dimensional visualization techniques combined with a quantitative analysis revealed that the events of final envelopment are equally distributed within the AC irrespective of different local membrane composition.


Assuntos
Citomegalovirus/ultraestrutura , Células Endoteliais/ultraestrutura , Fibroblastos/ultraestrutura , Macrófagos/ultraestrutura , Montagem de Vírus/fisiologia , Células Cultivadas , Citomegalovirus/química , Citomegalovirus/genética , Células Endoteliais/metabolismo , Células Endoteliais/virologia , Fibroblastos/metabolismo , Fibroblastos/virologia , Humanos , Imageamento Tridimensional , Macrófagos/metabolismo , Macrófagos/virologia , Microscopia Eletrônica , Microtomia , Mutação , Inclusão do Tecido , Proteínas Virais/genética , Proteínas Virais/metabolismo
20.
Traffic ; 13(11): 1443-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22805610

RESUMO

Enveloped viruses acquire their host-derived membrane at a variety of intracellular locations. Herpesviruses are complex entities that undergo several budding and fusion events during an infection. All members of this large family are believed to share a similar life cycle. However, they seemingly differ in terms of acquisition of their mature envelope. Herpes simplex virus is often believed to bud into an existing intracellular compartment, while the related cytomegalovirus may acquire its final envelope from a novel virus-induced assembly compartment. This review focuses on recent advances in the characterization of cellular compartment(s) potentially contributing to herpes virion final envelopment. It also examines the common points between seemingly distinct envelopment pathways and highlights the dynamic nature of intracellular compartments in the context of herpesvirus infections.


Assuntos
Herpesviridae/fisiologia , Internalização do Vírus , Animais , Capsídeo/fisiologia , Capsídeo/ultraestrutura , Citomegalovirus/metabolismo , Citomegalovirus/fisiologia , Citomegalovirus/ultraestrutura , Citosol/metabolismo , Citosol/virologia , Interações Hospedeiro-Patógeno , Organelas/metabolismo , Organelas/virologia , Proteínas do Envelope Viral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...