Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Virol ; 88(10): 5559-77, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24600013

RESUMEN

UNLABELLED: Epstein-Barr virus (EBV) attachment to human CD21 on the B-cell surface initiates infection. Whether CD21 is a simple tether or conveys vital information to the cell interior for production of host factors that promote infection of primary B cells is controversial, as the cytoplasmic fragment of CD21 is short, though highly conserved. The ubiquity of CD21 on normal B cells, the diversity of this population, and the well-known resistance of primary B cells to gene transfer technologies have all impeded resolution of this question. To uncover the role(s) of the CD21 cytoplasmic domain during infection initiation, the full-length receptor (CD21=CR), a mutant lacking the entire cytoplasmic tail (CT), and a control vector (NEO) were stably expressed in two pre-B-cell lines that lack endogenous receptor. Genome-wide transcriptional analysis demonstrated that stable CD21 surface expression alone (either CR or CT) produced multiple independent changes in gene expression, though both dramatically decreased class I melanoma-associated antigen (MAGE) family RNAs and upregulated genes associated with B-cell differentiation (e.g., C2TA, HLA-II, IL21R, MIC2, CD48, and PTPRCAP/CD45-associated protein). Temporal analysis spanning 72 h revealed that not only CR- but also CT-expressing lines initiated latency. In spite of this, the number and spectrum of transcripts altered in CR- compared with CT-bearing lines at 1 h after infection further diverged. Differential modulation of immediate early cellular transcripts (e.g., c-Jun and multiple histones), both novel and previously linked to CD21-initiated signaling, as well as distinct results from pathway analyses support a separate role for the cytoplasmic domain in initiation of intracellular signals. IMPORTANCE: Membrane proteins that mediate virus attachment tether virus particles to the cell surface, initiating infection. In addition, upon virus interaction such proteins may transmit signals to the interior of the cell that support subsequent steps in the infection process. Here we show that expression of the Epstein-Barr virus B-cell attachment receptor, CD21, in B cells that lack this receptor results in significant changes in gene expression, both before and rapidly following EBV-CD21 interaction. These changes translate into major signaling pathway alterations that are predicted to support stable infection.


Asunto(s)
Linfocitos B/fisiología , Linfocitos B/virología , Diferenciación Celular , Herpesvirus Humano 4/fisiología , Interacciones Huésped-Patógeno , Receptores de Complemento 3d/metabolismo , Acoplamiento Viral , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Estructura Terciaria de Proteína , Receptores de Complemento 3d/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal
2.
J Transl Med ; 13: 50, 2015 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-25885535

RESUMEN

Epstein-Barr virus (EBV), an oncogenic gammaherpesvirus, causes acute infectious mononucleosis (AIM) and is linked to the development of several human malignancies. There is an urgent need for a vaccine that is safe, prevents infection and/or limits disease. Unique among human herpesviruses, glycoprotein (gp)350/220, which initiates EBV attachment to susceptible host cells, is the major ligand on the EBV envelope and is highly conserved. Interaction between gp350/220 and complement receptor type 2 (CR2)/CD21 and/or (CR1)/CD35 on B-cells is required for infection. Potent antibody responses to gp350/220 occur in animal models and humans. Thus, gp350/220 provides an attractive candidate for prophylactic subunit vaccine development. However, in a recent Phase II clinical trial immunization with soluble recombinant gp350 reduced the incidence of AIM, but did not prevent infection. Despite various attempts to produce an EBV vaccine, no vaccine is licensed. Herein we describe a sub-unit vaccine against EBV based on a novel Newcastle disease virus (NDV)-virus-like particle (VLP) platform consisting of EBVgp350/220 ectodomain fused to NDV-fusion (F) protein. The chimeric protein EBVgp350/220-F is incorporated into the membrane of a VLP composed of the NDV matrix and nucleoprotein. The particles resemble native EBV in diameter and shape and bind CD21 and CD35. Immunization of BALB/c mice with EBVgp350/220-F VLPs elicited strong, long-lasting neutralizing antibody responses when assessed in vitro. This chimeric VLP is predicted to provide a superior safety profile as it is efficiently produced in Chinese hamster ovary (CHO) cells using a platform devoid of human nucleic acid and EBV-transforming genes.


Asunto(s)
Anticuerpos Neutralizantes/biosíntesis , Linfocitos B/citología , Proteínas Recombinantes/metabolismo , Proteínas de la Matriz Viral/inmunología , Virión/metabolismo , Animales , Antígenos CD/metabolismo , Adhesión Celular , Línea Celular , Humanos , Inmunización , Inmunoglobulina G/metabolismo , Ratones Endogámicos BALB C , Pruebas de Neutralización , Unión Proteica
3.
J Virol ; 87(18): 10126-38, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23843639

RESUMEN

All eight human herpesviruses have a conserved herpesvirus protein kinase (CHPK) that is important for the lytic phase of the viral life cycle. In this study, we show that heat shock protein 90 (Hsp90) interacts directly with each of the eight CHPKs, and we demonstrate that an Hsp90 inhibitor drug, 17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG), decreases expression of all eight CHPKs in transfected HeLa cells. 17-DMAG also decreases expression the of the endogenous Epstein-Barr virus protein kinase (EBV PK, encoded by the BGLF4 gene) in lytically infected EBV-positive cells and inhibits phosphorylation of several different known EBV PK target proteins. Furthermore, 17-DMAG treatment abrogates expression of the human cytomegalovirus (HCMV) kinase UL97 in HCMV-infected human fibroblasts. Importantly, 17-DMAG treatment decreased the EBV titer approximately 100-fold in lytically infected AGS-Akata cells without causing significant cellular toxicity during the same time frame. Increased EBV PK expression in 17-DMAG-treated AGS-Akata cells did not restore EBV titers, suggesting that 17-DMAG simultaneously targets multiple viral and/or cellular proteins required for efficient viral replication. These results suggest that Hsp90 inhibitors, including 17-DMAG, may be a promising group of drugs that could have profound antiviral effects on herpesviruses.


Asunto(s)
Antivirales/metabolismo , Benzoquinonas/metabolismo , Inhibidores Enzimáticos/metabolismo , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Herpesvirus Humano 4/fisiología , Lactamas Macrocíclicas/metabolismo , Proteínas Quinasas/metabolismo , Replicación Viral/efectos de los fármacos , Línea Celular , Células Epiteliales/efectos de los fármacos , Células Epiteliales/virología , Fibroblastos/efectos de los fármacos , Fibroblastos/virología , Herpesvirus Humano 4/efectos de los fármacos , Humanos , Mapeo de Interacción de Proteínas , Carga Viral , Cultivo de Virus
4.
J Immunol ; 188(9): 4496-505, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22490440

RESUMEN

Asplenic individuals are compromised not only in their ability to destroy infectious agents, but are at increased risk for death from autoimmune disease, certain tumors, and ischemic heart disease. Enhanced mortality is attributed to lack of phagocytes sequestered in spleen that efficiently engulf and destroy appropriate targets, although related cells are found elsewhere. To determine whether a unique population regulates RBC-pathogen clearance and filtration of altered self, we reviewed the anatomic literature and analyzed in situ by immunohistochemistry and immunofluorescence the expression patterns of a little-characterized cell that dominates the splenic red pulp of humans and closely related primates: the venous sinus-lining or littoral cell (LC). High expression of the formin homology domain protein 1 outlines the LC population. Although LCs are endothelial-like in distribution, they express several macrophage-directed proteins, the RBC Duffy Ag receptor for chemokines and T cell coreceptor CD8α/α, yet they lack lineage-associated markers CD34 and CD45. Strikingly, SIRPα (CD172a) expression in human spleen concentrates on LCs, consistent with recent demonstration of a key role in RBC turnover and elimination versus release of infected or altered self. Our results indicate human LCs (SIRPα(+), formin homology domain protein 1(+), CD8α/α(+), CD34(-), CD45(-)) comprise a highly plastic barrier cell population that emerged late in primate evolution coordinate with CD8 expression. Unique to Hominidae, LCs may be the ultimate determinant of which cells recirculate after passage through human spleen.


Asunto(s)
Antígenos de Diferenciación/inmunología , Proteínas Fetales/inmunología , Proteínas Nucleares/inmunología , Receptores Inmunológicos/inmunología , Bazo/inmunología , Antígenos CD/biosíntesis , Antígenos CD/inmunología , Antígenos de Diferenciación/metabolismo , Biomarcadores/metabolismo , Sistema del Grupo Sanguíneo Duffy/biosíntesis , Sistema del Grupo Sanguíneo Duffy/inmunología , Femenino , Proteínas Fetales/metabolismo , Forminas , Regulación de la Expresión Génica/inmunología , Humanos , Inmunohistoquímica , Masculino , Proteínas Nucleares/metabolismo , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/inmunología , Receptores Inmunológicos/metabolismo , Bazo/citología , Bazo/metabolismo
5.
Nat Genet ; 36(7): 683-5, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15220917

RESUMEN

Kaposi sarcoma is considered a neoplasm of lymphatic endothelium infected with Kaposi sarcoma-associated herpesvirus. It is characterized by the expression of lymphatic lineage-specific genes by Kaposi sarcoma tumor cells. Here we show that infection of differentiated blood vascular endothelial cells with Kaposi sarcoma-associated herpesvirus leads to their lymphatic reprogramming; induction of approximately 70% of the main lymphatic lineage-specific genes, including PROX1, a master regulator of lymphatic development; and downregulation of blood vascular genes.


Asunto(s)
Endotelio/patología , Herpesvirus Humano 8/fisiología , Vasos Linfáticos/patología , Células Cultivadas , Regulación hacia Abajo , Endotelio/metabolismo , Endotelio/virología , Perfilación de la Expresión Génica , Vasos Linfáticos/metabolismo , Vasos Linfáticos/virología
6.
Breast Cancer ; 29(4): 731-739, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35347575

RESUMEN

BACKGROUND: The link between Epstein-Barr Virus (EBV) and breast cancer (BC) etiology remains unclear. We utilized the Health of Women (HOW) Study® to understand the association between infectious mononucleosis (IM), a surrogate for EBV infection, and invasive BC. METHODS: The HOW Study® was a web-based survey of BC risk factors with > 40, 000 participants; 183 had IM at < 10 years old, 3, 654 had IM between 10 and 22 years old, 764 had IM at > 22 years old, and 17, 026 never developed IM. Of these 21, 627 women, 2093 had Stages I-III BC and 14, 143 were cancer-free. Binary logistic regression ascertained the association between IM and invasive BC risk by controlling for confounders. RESULTS: A history of IM was associated with a lower likelihood of developing invasive BC compared to women who did not develop IM (adjusted OR = 0.83, 95% CI 0.72-0.94). That finding was driven by women who had IM between 10 and 22 years old (adjusted OR = 0.83, 95% CI 0.72-0.97) albeit no linear association between age at developing IM and breast cancer (p-trend > 0.05). Women who had IM between 10 and 22 years old were less likely to develop estrogen receptor positive (ER+ ; adjusted OR = 0.84, 95% CI 0.71-0.99) or hormone receptor positive (HR+ ; adjusted OR = 0.86, 95% CI 0.73-1.01) BC. There was no association between IM and ER- or HR- BC. CONCLUSION: In the HOW Study®, women diagnosed with IM between the ages of 10 and 22 had a lower risk of developing invasive BC compared to women who never developed IM.


Asunto(s)
Neoplasias de la Mama , Infecciones por Virus de Epstein-Barr , Mononucleosis Infecciosa , Adolescente , Adulto , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/etiología , Niño , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/diagnóstico , Femenino , Herpesvirus Humano 4 , Humanos , Mononucleosis Infecciosa/complicaciones , Mononucleosis Infecciosa/epidemiología , Modelos Logísticos , Adulto Joven
7.
J Virol ; 84(9): 4534-42, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20181711

RESUMEN

Ganciclovir (GCV) and acyclovir (ACV) are guanine nucleoside analogues that inhibit lytic herpesvirus replication. GCV and ACV must be monophosphorylated by virally encoded enzymes to be converted into nucleotides and incorporated into viral DNA. However, whether GCV and/or ACV phosphorylation in Epstein-Barr virus (EBV)-infected cells is mediated primarily by the EBV-encoded protein kinase (EBV-PK), the EBV-encoded thymidine kinase (EBV-TK), or both is controversial. To examine this question, we constructed EBV mutants containing stop codons in either the EBV-PK or EBV-TK open reading frame and selected for stable 293T clones latently infected with wild-type EBV or each of the mutant viruses. Cells were induced to the lytic form of viral replication with a BZLF1 expression vector in the presence and absence of various doses of GCV and ACV, and infectious viral titers were determined by a green Raji cell assay. As expected, virus production in wild-type EBV-infected 293T cells was inhibited by both GCV (50% inhibitory concentration [IC(50)] = 1.5 microM) and ACV (IC(50) = 4.1 microM). However, the EBV-PK mutant (which replicates as well as the wild-type (WT) virus in 293T cells) was resistant to both GCV (IC(50) = 19.6 microM) and ACV (IC(50) = 36.4 microM). Expression of the EBV-PK protein in trans restored GCV and ACV sensitivity in cells infected with the PK mutant virus. In contrast, in 293T cells infected with the TK mutant virus, viral replication remained sensitive to both GCV (IC(50) = 1.2 microM) and ACV (IC(50) = 2.8 microM), although susceptibility to the thymine nucleoside analogue, bromodeoxyuridine, was reduced. Thus, EBV-PK but not EBV-TK mediates ACV and GCV susceptibilities.


Asunto(s)
Aciclovir/farmacología , Antivirales/farmacología , Ganciclovir/farmacología , Herpesvirus Humano 4/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Timidina Quinasa/metabolismo , Proteínas Virales/metabolismo , Aciclovir/metabolismo , Antivirales/metabolismo , Línea Celular , Codón sin Sentido , Ganciclovir/metabolismo , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/crecimiento & desarrollo , Humanos , Concentración 50 Inhibidora , Mutagénesis Sitio-Dirigida , Fosforilación , Proteínas Serina-Treonina Quinasas/deficiencia , Timidina Quinasa/deficiencia , Proteínas Virales/genética , Replicación Viral/efectos de los fármacos
8.
EBioMedicine ; 9: 148-160, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27333046

RESUMEN

Whether the human tumor virus, Epstein-Barr Virus (EBV), promotes breast cancer remains controversial and a potential mechanism has remained elusive. Here we show that EBV can infect primary mammary epithelial cells (MECs) that express the receptor CD21. EBV infection leads to the expansion of early MEC progenitor cells with a stem cell phenotype, activates MET signaling and enforces a differentiation block. When MECs were implanted as xenografts, EBV infection cooperated with activated Ras and accelerated the formation of breast cancer. Infection in EBV-related tumors was of a latency type II pattern, similar to nasopharyngeal carcinoma (NPC). A human gene expression signature for MECs infected with EBV, termed EBVness, was associated with high grade, estrogen-receptor-negative status, p53 mutation and poor survival. In 11/33 EBVness-positive tumors, EBV-DNA was detected by fluorescent in situ hybridization for the viral LMP1 and BXLF2 genes. In an analysis of the TCGA breast cancer data EBVness correlated with the presence of the APOBEC mutational signature. We conclude that a contribution of EBV to breast cancer etiology is plausible, through a mechanism in which EBV infection predisposes mammary epithelial cells to malignant transformation, but is no longer required once malignant transformation has occurred.


Asunto(s)
Transformación Celular Neoplásica , Herpesvirus Humano 4/patogenicidad , Neoplasias/patología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Células Cultivadas , Análisis por Conglomerados , ADN Viral/genética , ADN Viral/metabolismo , Supervivencia sin Enfermedad , Células Epiteliales/citología , Células Epiteliales/trasplante , Células Epiteliales/virología , Transición Epitelial-Mesenquimal , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Humanos , Immunoblotting , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neoplasias/metabolismo , Neoplasias/mortalidad , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Complemento 3d/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Tasa de Supervivencia , Transcriptoma , Trasplante Heterólogo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas de la Matriz Viral/antagonistas & inhibidores , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo
9.
Cell Rep ; 3(2): 371-85, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-23416052

RESUMEN

Epstein-Barr virus (EBV) attachment to primary B cells initiates virus entry. Although CD21 is the only known receptor for EBVgp350/220, a recent report documents EBV-infected B cells from a patient genetically deficient in CD21. On normal resting B cells, CD21 forms two membrane complexes: one with CD19 and another with CD35. Whereas the CD21/CD19 complex is widely retained on immortalized and B cell tumor lines, the related complement-regulatory protein CD35 is lost. To determine the role(s) of CD35 in initial infection, we transduced a CD21-negative pre-B cell and myeloid leukemia line with CD35, CD21, or both. Cells expressing CD35 alone bound gp350/220 and became latently infected when the fusion receptor HLA II was coexpressed. Temporal, biophysical, and structural characteristics of CD35-mediated infection were distinct from CD21. Identification of CD35 as an EBV receptor uncovers a salient role in primary infection, addresses unsettled questions of virus tropism, and underscores the importance of EBVgp350/220 for vaccine development.


Asunto(s)
Herpesvirus Humano 4/metabolismo , Receptores de Complemento 3b/metabolismo , Receptores de Complemento 3d/metabolismo , Antígenos CD19/metabolismo , Línea Celular , Humanos , Células K562 , Mutación , Células Precursoras de Linfocitos B/inmunología , Células Precursoras de Linfocitos B/metabolismo , Unión Proteica , Receptores de Complemento 3b/genética , Receptores de Complemento 3d/genética , Temperatura , Transfección , Proteínas de la Matriz Viral/metabolismo , Acoplamiento Viral , Internalización del Virus
10.
J Virol ; 81(12): 6523-35, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17428875

RESUMEN

The thymidine kinase (TK) encoded by Epstein-Barr virus (EBV) differs not only from that of the alphaherpesviruses but also from that of the gamma-2 herpesvirus subfamily. Because cellular location is frequently a determinant of regulatory function, to gain insight into additional role(s) of EBV TK and to uncover how the lymphocryptovirus and rhadinovirus enzymes differ, the subcellular localizations of EBV TK and the related cercopithecine herpesvirus-15 TK were investigated. We show that in contrast to those of the other family members, the gamma-1 herpesvirus TKs localize to the centrosome and even more precisely to the periphery of the centriole, tightly encircling the tubulin-rich centrioles in a microtubule-independent fashion. Centrosomal localization is observed in diverse cell types and occurs whether the protein is expressed independently or in the context of lytic EBV infection. Surprisingly, analysis of mutants revealed that the unique N-terminal domain was not critical for targeting to the centrosome, but rather, peptide sequences located C terminal to this domain were key. This is the first herpesvirus protein documented to reside in the centrosome, or microtubule-organizing center, an amembranous organelle that regulates the structural biology of the cell cycle through control of chromosome separation and cytokinesis. More recently, proteasome-mediated degradation of cell cycle regulatory proteins, production and loading of antigenic peptides onto HLA molecules, and transient homing of diverse virion proteins required for entry and/or egress have been shown to be coordinated at the centrosome. Potential implications of centrosomal localization for EBV TK function are discussed.


Asunto(s)
Centriolos/virología , Herpesvirus Humano 4/enzimología , Timidina Quinasa/fisiología , Secuencia de Aminoácidos , Animales , Células CHO , Ciclo Celular , Centrosoma/metabolismo , Centrosoma/ultraestructura , Cricetinae , Cricetulus , Humanos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Timidina Quinasa/biosíntesis , Tubulina (Proteína)/química
11.
J Virol ; 79(23): 14647-59, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16282465

RESUMEN

The nucleoside kinase encoded by Kaposi's sarcoma-associated herpesvirus (KSHV) is a relatively inefficient enzyme with substrate specificity for thymidine alone, unlike alphaherpesvirus thymidine kinases (TKs). Similar to all gammaherpesvirus TKs, KSHV TK is composed of two distinct domains, a conserved C-terminal kinase and a novel and uncharacterized N terminus. Ectopic expression of KSHV TK in adherent cells induced striking morphological changes and anchorage independence although cells survived, a property shared with the related rhadinovirus TKs of rhesus monkey rhadinovirus and herpesvirus saimiri. To determine whether KSHV TK served alternate functions relevant to the rhadinovirus life cycle and to reveal the contribution of the N terminus, an enhanced green fluorescent protein-tagged fusion protein and serial mutants were generated for investigation of intracellular localization and cell biology. Analysis of truncation mutants showed that a proline-rich region located within the N terminus cooperated with the conserved C-terminal kinase to tether KSHV TK to a reticular network in the cytoplasm and to induce morphological change. Fusion of the KSHV N terminus to herpes simplex virus type 1 TK, a nucleus-localized enzyme, similarly resulted in cytoplasmic redistribution of the chimeric protein but did not alter cell shape or adhesion. Unlike other human herpesvirus TKs, KSHV TKs and related rhadinovirus TKs are constitutively tyrosine phosphorylated; a KSHV TK mutant that was hypophosphorylated failed to detach and grow in suspension. Loss of adhesion may enhance terminal differentiation, viral replication, and egress at the cellular level and at the organism level may facilitate detachment and distant migration of KSHV-replicating cells within body fluids--promoting oropharyngeal transmission and perhaps contributing to the multifocal lesions that characterize KS.


Asunto(s)
Herpesvirus Humano 8/fisiología , Fosfoproteínas/farmacología , Rhadinovirus/fisiología , Sarcoma de Kaposi/virología , Timidina Quinasa/metabolismo , Adhesión Celular/efectos de los fármacos , Técnicas de Cultivo de Célula , Herpesvirus Humano 8/metabolismo , Humanos , Rhadinovirus/metabolismo , Sarcoma de Kaposi/patología
12.
Blood ; 100(3): 888-96, 2002 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12130499

RESUMEN

Kaposi sarcoma-associated herpesvirus (KSHV)/human herpesvirus-8 (HHV-8) is causally associated with Kaposi sarcoma (KS). The absence of a cell culture system that effectively reproduces the composite mechanisms governing initiation and maintenance of HHV-8 infection (lytic and latent) in KS endothelial cells, however, has left important questions unanswered. Here, we report a culture system in which the earliest events that accompany HHV-8 infection could be surveyed in primary endothelial cells. Binding of HHV-8 to microvascular dermal endothelial cells (MVDECs) was directly compared with other primary target cells implicated in HHV-8-associated diseases. Virus attachment, fusion, internalization and transport within MVDECs was monitored by electron microscopy. Studies of genome configuration revealed that rapid circularization of the viral DNA occurred on entry, though by 72 hours after infection linear DNAs accumulated and early as well as late lytic RNAs (T1.1, K8.1) could be detected. The latency transcripts (LT1/LT2) were first detected on day 8, demonstrating that both lytic and latent infection were initiated. Although most lytic transcripts accrued until passage, open-reading frame-74 RNAs fluctuated with a fixed periodicity, suggesting that early replication after infection of MVDECs was synchronous.


Asunto(s)
Endotelio Vascular/virología , Infecciones por Herpesviridae , Sarcoma de Kaposi/virología , Técnicas de Cultivo de Célula/métodos , ADN Viral/metabolismo , ADN Viral/fisiología , ADN Viral/ultraestructura , Endotelio Vascular/citología , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/crecimiento & desarrollo , Humanos , Microscopía Electrónica , Hibridación de Ácido Nucleico , ARN Viral/metabolismo , ARN Viral/fisiología , ARN Viral/ultraestructura , Factores de Tiempo , Células Tumorales Cultivadas , Cultivo de Virus , Replicación Viral
13.
Virology ; 299(1): 109-21, 2002 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-12167346

RESUMEN

Epstein-Barr virus (EBV) encodes multiple latency programs: a growth-transforming program (type III) latency program and restricted-latency (types I and II) programs. During type III latency, EBV expresses six nuclear antigens, all of which are encoded by a single complex transcriptional unit driven by two linked promoters, Cp and Wp, while restricted viral latency is characterized by the expression of a single nuclear antigen, EBNA1, whose expression is driven from a distinct transcription unit under the control of the Qp promoter. EBV infection of the 293 epithelial cell line frequently leads to the establishment of a type I/II latent infection. Here we report that during the initial stages of virus infection of the 293 cell line, both Cp and Wp are active. However, analysis of four established, low-passage EBV-infected 293 cell lines revealed that three of these exhibited Qp-driven transcription of the EBNA 1 gene and little or no detectable Cp and Wp activity, while the fourth cell line exhibited Cp activity. Notably, all four cell lines contained the necessary transcription factors to drive transcription initiation from Cp and Wp when transiently transfected with unmethylated reporter constructs. Furthermore, in the cell lines exhibiting restricted EBV latency the viral genomes were extensively methylated around Cp and Wp, but not Qp. In contrast, in the cell line exhibiting Cp activity the viral genomes were hypomethylated around Cp, Wp, and Qp. Taken together, these results provide evidence that the establishment of a restricted latent infection in the 293 epithelial cell line is not due to a failure to initiate the growth-transforming (type III) latency program, but rather may arise from a selection against the type III latency program. Furthermore, these results are consistent with the hypothesis that methylation of Cp and Wp is required for entry into the type I or II latency programs.


Asunto(s)
Metilación de ADN , ADN Viral/genética , Células Epiteliales/virología , Herpesvirus Humano 4/fisiología , Latencia del Virus/genética , Línea Celular Transformada , Antígenos Nucleares del Virus de Epstein-Barr/genética , Herpesvirus Humano 4/genética , Humanos , Riñón , Regiones Promotoras Genéticas , Transcripción Genética
14.
Proc Natl Acad Sci U S A ; 99(16): 10641-6, 2002 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-12122212

RESUMEN

Human complement receptor type 2 (CD21) is the cellular receptor for Epstein-Barr virus (EBV), a human tumor virus. The N-terminal two short consensus repeats (SCR1-SCR2) of the receptor interact with the EBV glycoprotein gp350/220 and also with the natural CD21 ligand C3d. Here we present the crystal structure of the CD21 SCR1-SCR2 fragment in the absence of ligand and demonstrate that it is able to bind EBV. Based on a functional analysis of wild-type and mutant CD21 and molecular modeling, we identify a likely region for EBV attachment and demonstrate that this region is not involved in the interaction with C3d. A comparison with the previously determined structure of CD21 SCR1-SCR2 in complex with C3d shows that, in both cases, CD21 assumes compact V-shaped conformations. However, our analysis reveals a surprising degree of flexibility at the SCR1-SCR2 interface, suggesting interactions between the two domains are not specific. We present evidence that the V-shaped conformation is induced by deglycosylation of the protein, and that physiologic glycosylation of CD21 would result in a more extended conformation, perhaps with additional epitopes for C3d binding.


Asunto(s)
Complemento C3d/química , Herpesvirus Humano 4/química , Receptores de Complemento 3d/química , Secuencia de Carbohidratos , Complemento C3d/inmunología , Cristalografía por Rayos X , Herpesvirus Humano 4/inmunología , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Receptores de Complemento 3d/genética , Receptores de Complemento 3d/inmunología
15.
J Cell Sci ; 117(Pt 13): 2709-20, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15138285

RESUMEN

CD21 is a multifunctional receptor for Epstein-Barr virus (EBV), for C3dg and for CD23. Upon engagement of immune complexes CD21 modulates immunoreceptor signaling, linking innate and adaptive immune responses. The mechanisms enabling CD21 to independently relay information between the exterior and interior of the cell, however, remain unresolved. We show that formin homologue overexpressed in spleen (FHOS/FHOD1) binds the cytoplasmic domain of human CD21 through its C terminus. When expressed in cells, EGFP-FHOS localizes to the cytoplasm and accumulates with actin in membrane protrusions. Plasma membrane aggregation, redistribution and co-localization of both proteins are stimulated when EBV (ligand) binds CD21. Though widely expressed, FHOS RNA is most abundant in the littoral cell, a major constituent of the red pulp of human spleen believed to function in antigen filtration. Formins are molecular scaffolds that nucleate actin by a pathway distinct from Arp2/3 complex, linking signal transduction to actin reorganization and gene transcription. Thus, ligand stimulation of FHOS-CD21 interaction may transmit signals through promotion of cytoskeletal rearrangement. Moreover, formin recruitment to sites of actin assembly initiated by immunoreceptors could be a general mechanism whereby co-receptors such as CD21 modulate intracellular signaling.


Asunto(s)
Proteínas Fetales/metabolismo , Herpesvirus Humano 4/metabolismo , Proteínas Nucleares/metabolismo , Receptores de Complemento 3d/química , Receptores de Complemento 3d/metabolismo , Células 3T3 , Adenoviridae/genética , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Línea Celular Transformada , Línea Celular Tumoral , Transformación Celular Viral , Citoplasma/química , Citoplasma/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Forminas , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Ratones , Microscopía Fluorescente , Modelos Biológicos , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA