Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Virol ; 95(5)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33328302

RESUMEN

The CD200-CD200R pathway is involved in inhibition of immune responses, and the importance of this pathway to infectious disease is highlighted by the fact that viral CD200 (vCD200) molecules have been found to be encoded by several DNA viruses, including the human gammaherpesvirus Kaposi's sarcoma-associated herpesvirus (KSHV), and the closely related rhesus macaque rhadinovirus (RRV). KSHV vCD200 is the most extensively studied vCD200 molecule, however, the only herpesvirus vCD200 molecule to be examined in vivo is that encoded by RRV. Our prior studies have demonstrated that RRV vCD200 is a functional CD200 homologue that is capable of affecting immune responses in vivo, and further, that RRV can express a secreted form of vCD200 (vCD200-Sec) during infection. Despite this information, RRV vCD200 has not been examined specifically for effects on RM CD200R signaling, and the functionality of vCD200-Sec has not been examined in any context. Thus, we developed an in vitro model system in which B cells expressing vCD200 were utilized to assess the effects of this molecule on the regulation of myeloid cells expressing RM CD200R, mimicking interactions that are predicted to occur in vivo Our findings suggest that RRV vCD200 can bind and induce functional signals through RM CD200R, while vCD200-Sec represents a non-functional protein incapable of affecting CD200R signaling. We also provide the first demonstration of the function of RM CD200, which appears to possess more robust signaling capabilities than RRV vCD200, and also show that KSHV vCD200 does not efficiently induce signaling via RM CD200R.IMPORTANCE Viral CD200 homologues are encoded by KSHV and the closely related RRV. Though RRV vCD200 has been examined, questions still exist in regard to the ability of this molecule to induce signaling via rhesus macaque CD200R, as well as the potential function of a secreted form of vCD200. Further, all previous in vitro studies of RRV vCD200 have utilized an Fc fusion protein to examine functionality, which does not replicate the structural properties of the membrane-associated form of vCD200 that is naturally produced during RRV infection. In this study, we demonstrate for the first time that membrane-expressed RRV vCD200 is capable of inducing signal transduction via RM CD200R, while the secreted form of vCD200 appears to be non-functional. Further, we also demonstrate that RM CD200 induces signaling via RM CD200R, and is more robust than RRV vCD200, while KSHV vCD200 does not appear to induce efficient signaling via RM CD200R.

2.
J Virol ; 93(6)2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30626678

RESUMEN

Interferon (IFN) production and the subsequent induction of IFN-stimulated genes (ISGs) are highly effective innate strategies utilized by cells to protect against invading pathogens, including viruses. Critical components involved in this innate process are promyelocytic leukemia nuclear bodies (PML-NBs), which are subnuclear structures required for the development of a robust IFN response. As such, PML-NBs serve as an important hurdle for viruses to overcome to successfully establish an infection. Both Kaposi's sarcoma-associated herpesvirus (KSHV) and the closely related rhesus macaque rhadinovirus (RRV) are unique for encoding viral homologs of IFN regulatory factors (termed vIRFs) that can manipulate the host immune response by multiple mechanisms. All four KSHV vIRFs inhibit the induction of IFN, while vIRF1 and vIRF2 can inhibit ISG induction downstream of the IFN receptor. Less is known about the RRV vIRFs. RRV vIRF R6 can inhibit the induction of IFN by IRF3; however, it is not known whether any RRV vIRFs inhibit ISG induction following IFN receptor signaling. In our present study, we demonstrate that the RRV vIRF R12 aids viral replication in the presence of the type I IFN response. This is achieved in part through the disruption of PML-NBs and the inhibition of robust ISG transcription.IMPORTANCE KSHV and RRV encode a unique set of homologs of cellular IFN regulatory factors, termed vIRFs, which are hypothesized to help these viruses evade the innate immune response and establish infections in their respective hosts. Our work elucidates the role of one RRV vIRF, R12, and demonstrates that RRV can dampen the type I IFN response downstream of IFN signaling, which would be important for establishing a successful infection in vivo.


Asunto(s)
Factores Reguladores del Interferón/genética , Interferón Tipo I/genética , Cuerpos de Inclusión Intranucleares/genética , Leucemia Promielocítica Aguda/genética , Macaca mulatta/virología , Rhadinovirus/genética , Transducción de Señal/genética , Proteínas Virales/genética , Animales , Línea Celular , Herpesvirus Humano 8/genética , Humanos , Inmunidad Innata/genética , Factor 3 Regulador del Interferón/genética , Leucemia Promielocítica Aguda/virología , Receptores de Interferón/genética , Transcripción Genética/genética , Replicación Viral/genética
3.
PLoS Negl Trop Dis ; 9(6): e0003850, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26086739

RESUMEN

Monkeypox virus (MPXV) is the etiological agent of human (MPX). It is an emerging orthopoxvirus zoonosis in the tropical rain forest of Africa and is endemic in the Congo-basin and sporadic in West Africa; it remains a tropical neglected disease of persons in impoverished rural areas. Interaction of the human population with wildlife increases human infection with MPX virus (MPXV), and infection from human to human is possible. Smallpox vaccination provides good cross-protection against MPX; however, the vaccination campaign ended in Africa in 1980, meaning that a large proportion of the population is currently unprotected against MPXV infection. Disease control hinges on deterring zoonotic exposure to the virus and, barring that, interrupting person-to-person spread. However, there are no FDA-approved therapies against MPX, and current vaccines are limited due to safety concerns. For this reason, new studies on pathogenesis, prophylaxis and therapeutics are still of great interest, not only for the scientific community but also for the governments concerned that MPXV could be used as a bioterror agent. In the present study, a new vaccination strategy approach based on three recombinant bovine herpesvirus 4 (BoHV-4) vectors, each expressing different MPXV glycoproteins, A29L, M1R and B6R were investigated in terms of protection from a lethal MPXV challenge in STAT1 knockout mice. BoHV-4-A-CMV-A29LgD106ΔTK, BoHV-4-A-EF1α-M1RgD106ΔTK and BoHV-4-A-EF1α-B6RgD106ΔTK were successfully constructed by recombineering, and their capacity to express their transgene was demonstrated. A small challenge study was performed, and all three recombinant BoHV-4 appeared safe (no weight-loss or obvious adverse events) following intraperitoneal administration. Further, BoHV-4-A-EF1α-M1RgD106ΔTK alone or in combination with BoHV-4-A-CMV-A29LgD106ΔTK and BoHV-4-A-EF1α-B6RgD106ΔTK, was shown to be able to protect, 100% alone and 80% in combination, STAT1(-/-) mice against mortality and morbidity. This work demonstrated the efficacy of BoHV-4 based vectors and the use of BoHV-4 as a vaccine-vector platform.


Asunto(s)
Antígenos Virales/inmunología , Herpesvirus Bovino 4/fisiología , Monkeypox virus/inmunología , Mpox/prevención & control , Factor de Transcripción STAT1/metabolismo , Vacunas Virales/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Bovinos , Línea Celular , Regulación de la Expresión Génica , Vectores Genéticos , Herpesvirus Bovino 4/inmunología , Humanos , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Factor de Transcripción STAT1/genética , Transfección , Vacunas Virales/genética
4.
J Virol ; 88(18): 10635-54, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24991004

RESUMEN

UNLABELLED: Rhesus macaque rhadinovirus (RRV) is a gammaherpesvirus of rhesus macaque (RM) monkeys that is closely related to human herpesvirus 8 (HHV-8)/Kaposi's Sarcoma-associated herpesvirus (KSHV), and it is capable of inducing diseases in simian immunodeficiency virus (SIV)-infected RM that are similar to those seen in humans coinfected with HIV and HHV-8. Both HHV-8 and RRV encode viral CD200 (vCD200) molecules that are homologues of cellular CD200, a membrane glycoprotein that regulates immune responses and helps maintain immune homeostasis via interactions with the CD200 receptor (CD200R). Though the functions of RRV and HHV-8 vCD200 molecules have been examined in vitro, the precise roles that these viral proteins play during in vivo infection remain unknown. Thus, to address the contributions of RRV vCD200 to immune regulation and disease in vivo, we generated a form of RRV that lacked expression of vCD200 for use in infection studies in RM. Our data indicated that RRV vCD200 expression limits immune responses against RRV at early times postinfection and also impacts viral loads, but it does not appear to have significant effects on disease development. Further, examination of the distribution pattern of CD200R in RM indicated that this receptor is expressed on a majority of cells in peripheral blood mononuclear cells, including B and T cells, suggesting potentially wider regulatory capabilities for both vCD200 and CD200 that are not strictly limited to myeloid lineage cells. In addition, we also demonstrate that RRV infection affects CD200R expression levels in vivo, although vCD200 expression does not play a role in this phenomenon. IMPORTANCE: Cellular CD200 and its receptor, CD200R, compose a pathway that is important in regulating immune responses and is known to play a role in a variety of human diseases. A number of pathogens have been found to modulate the CD200-CD200R pathway during infection, including human herpesvirus 8 (HHV-8), the causative agent of Kaposi's sarcoma and B cell neoplasms in AIDS patients, and a closely related primate virus, rhesus macaque rhadinovirus (RRV), which infects and induces disease in rhesus macaque monkeys. HHV-8 and RRV encode homologues of CD200, termed vCD200, which are thought to play a role in preventing immune responses against these viruses. However, neither molecule has been studied in an in vivo model of infection to address their actual contributions to immunoregulation and disease. Here we report findings from our studies in which we analyzed the properties of a mutant form of RRV that lacks vCD200 expression in infected rhesus macaques.


Asunto(s)
Antígenos CD/inmunología , Infecciones por Herpesviridae/veterinaria , Enfermedades de los Monos/inmunología , Rhadinovirus/inmunología , Carga Viral , Proteínas Virales/inmunología , Animales , Antígenos CD/genética , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/virología , Macaca mulatta , Enfermedades de los Monos/genética , Enfermedades de los Monos/virología , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Rhadinovirus/genética , Rhadinovirus/fisiología , Proteínas Virales/genética
5.
Curr Opin Virol ; 3(3): 245-50, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23747119

RESUMEN

Rhesus macaque rhadinovirus (RRV) is a gamma-2 herpesvirus that naturally infects rhesus macaque (RM) monkeys and is closely related to human herpesvirus-8 (HHV-8)/Kaposi's sarcoma-associated herpesvirus (KSHV). Infection of immunodeficient RM induces disease in infected RM that resembles KSHV-associated pathologies. Importantly, RRV possesses homologues of KSHV ORFs that are postulated to play a role in disease development. As such, RRV has emerged as a prominent in vivo model system for examining mechanisms of infection and disease of these pathogenic herpesviruses, and has provided unique insight into how these viruses cause disease.


Asunto(s)
Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Enfermedades de los Primates/patología , Enfermedades de los Primates/virología , Rhadinovirus/patogenicidad , Infecciones Tumorales por Virus/patología , Infecciones Tumorales por Virus/virología , Animales , Macaca mulatta
6.
J Virol ; 87(1): 512-23, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23097433

RESUMEN

Japanese macaque rhadinovirus (JMRV) is a novel gamma-2 herpesvirus that was isolated from a Japanese macaque (JM) with an inflammatory demyelinating encephalomyelitis referred to as Japanese macaque encephalomyelitis, a disease that possesses clinical and histopathological features resembling multiple sclerosis in humans. Genomic DNA sequence analysis reveals that JMRV is a gammaherpesvirus closely related to rhesus macaque rhadinovirus (RRV) and human herpesvirus 8. We describe here the complete nucleotide sequence and structure of the JMRV genome, as well as the sequence of two plaque isolates of this virus. Analysis of the JMRV genome not only demonstrates that this virus shares a number of genes with RRV that may be involved in pathogenesis but also indicates the presence of unique JMRV genes that could potentially contribute to disease development. The knowledge of the genomic sequence of JMRV, and the ability to easily propagate the virus in vitro, make JMRV infection of JM an attractive model for examining the potential role of an infectious viral agent in the development of demyelinating encephalomyelitis disease in vivo.


Asunto(s)
ADN Viral/genética , Enfermedades Desmielinizantes/veterinaria , Encefalomielitis/veterinaria , Infecciones por Herpesviridae/veterinaria , Enfermedades de los Primates/virología , Rhadinovirus/genética , Animales , Análisis por Conglomerados , Enfermedades Desmielinizantes/virología , Encefalomielitis/virología , Genes Virales , Genoma Viral , Infecciones por Herpesviridae/virología , Macaca , Datos de Secuencia Molecular , Filogenia , Rhadinovirus/aislamiento & purificación , Análisis de Secuencia de ADN
7.
J Virol ; 86(5): 2769-79, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22171275

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) and the closely related gamma-2 herpesvirus rhesus macaque (RM) rhadinovirus (RRV) are the only known viruses to encode viral homologues of the cellular interferon (IFN) regulatory factors (IRFs). Recent characterization of a viral IRF (vIRF) deletion clone of RRV (vIRF-knockout RRV [vIRF-ko RRV]) demonstrated that vIRFs inhibit induction of type I and type II IFNs during RRV infection of peripheral blood mononuclear cells. Because the IFN response is a key component to a host's antiviral defenses, this study has investigated the role of vIRFs in viral replication and the development of the immune response during in vivo infection in RMs, the natural host of RRV. Experimental infection of RMs with vIRF-ko RRV resulted in decreased viral loads and diminished B cell hyperplasia, a characteristic pathology during acute RRV infection that often develops into more severe lymphoproliferative disorders in immune-compromised animals, similar to pathologies in KSHV-infected individuals. Moreover, in vivo infection with vIRF-ko RRV resulted in earlier and sustained production of proinflammatory cytokines and earlier induction of an anti-RRV T cell response compared to wild-type RRV infection. These findings reveal the broad impact that vIRFs have on pathogenesis and the immune response in vivo and are the first to validate the importance of vIRFs during de novo infection in the host.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por Herpesviridae/inmunología , Factores Reguladores del Interferón/inmunología , Macaca mulatta , Rhadinovirus/inmunología , Proteínas Virales/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/virología , Infecciones por Herpesviridae/virología , Humanos , Factores Reguladores del Interferón/genética , Interferones/inmunología , Rhadinovirus/genética , Proteínas Virales/genética
8.
J Virol ; 86(4): 2197-211, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22156526

RESUMEN

Kaposi's sarcoma-associated herpesvirus and rhesus macaque rhadinovirus (RRV), two closely related gammaherpesviruses, are unique in their expression of viral homologs of cellular interferon regulatory factors (IRFs), termed viral IRFs (vIRFs). To assess the role of vIRFs during de novo infection, we have utilized the bacterial artificial chromosome clone of wild-type RRV(17577) (WT(BAC) RRV) to generate a recombinant virus with all 8 of the vIRFs deleted (vIRF-ko RRV). The infection of primary rhesus fibroblasts and peripheral blood mononuclear cells (PBMCs) with vIRF-ko RRV resulted in earlier and increased induction of type I interferon (IFN) (IFN-α/ß) and type II IFN (IFN-γ). Additionally, plasmacytoid dendritic cells maintained higher levels of IFN-α production in PBMC cultures infected with vIRF-ko RRV than in cultures infected with WT(BAC) RRV. Moreover, the nuclear accumulation of phosphorylated IRF-3, which is necessary for the induction of type I IFN, was also inhibited following WT(BAC) RRV infection. These findings demonstrate that during de novo RRV infection, vIRFs are inhibiting the induction of IFN at the transcriptional level, and one potential mechanism for this is the disruption of the activation and localization of IRF-3.


Asunto(s)
Regulación hacia Abajo , Infecciones por Herpesviridae/veterinaria , Factores Reguladores del Interferón/metabolismo , Interferón Tipo I/genética , Interferón gamma/genética , Enfermedades de los Primates/genética , Rhadinovirus/fisiología , Proteínas Virales/metabolismo , Animales , Línea Celular , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/virología , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Factores Reguladores del Interferón/genética , Interferón Tipo I/metabolismo , Interferón gamma/metabolismo , Macaca mulatta , Enfermedades de los Primates/metabolismo , Enfermedades de los Primates/virología , Rhadinovirus/genética , Proteínas Virales/genética
9.
J Virol ; 85(18): 9527-42, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21752919

RESUMEN

Monkeypox virus (MPXV) is an orthopoxvirus closely related to variola virus, the causative agent of smallpox. Human MPXV infection results in a disease that is similar to smallpox and can also be fatal. Two clades of MPXV have been identified, with viruses of the central African clade displaying more pathogenic properties than those within the west African clade. The monkeypox inhibitor of complement enzymes (MOPICE), which is not expressed by viruses of the west African clade, has been hypothesized to be a main virulence factor responsible for increased pathogenic properties of central African strains of MPXV. To gain a better understanding of the role of MOPICE during MPXV-mediated disease, we compared the host adaptive immune response and disease severity following intrabronchial infection with MPXV-Zaire (n = 4), or a recombinant MPXV-Zaire (n = 4) lacking expression of MOPICE in rhesus macaques (RM). Data presented here demonstrate that infection of RM with MPXV leads to significant viral replication in the peripheral blood and lungs and results in the induction of a robust and sustained adaptive immune response against the virus. More importantly, we show that the loss of MOPICE expression results in enhanced viral replication in vivo, as well as a dampened adaptive immune response against MPXV. Taken together, these findings suggest that MOPICE modulates the anti-MPXV immune response and that this protein is not the sole virulence factor of the central African clade of MPXV.


Asunto(s)
Monkeypox virus/inmunología , Monkeypox virus/patogenicidad , Mpox/inmunología , Mpox/patología , Proteínas Virales/metabolismo , Factores de Virulencia/metabolismo , Inmunidad Adaptativa , Animales , Linfocitos B/inmunología , Sangre/virología , ADN Viral/química , ADN Viral/genética , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Pulmón/virología , Macaca mulatta , Masculino , Datos de Secuencia Molecular , Mpox/virología , Enfermedades de los Primates/inmunología , Enfermedades de los Primates/patología , Enfermedades de los Primates/virología , Análisis de Secuencia de ADN , Piel/patología , Linfocitos T/inmunología , Proteínas Virales/genética , Factores de Virulencia/genética
10.
Mol Cell Proteomics ; 9(12): 2760-71, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20736407

RESUMEN

Understanding viral pathogenesis is challenging because of confounding factors, including nonabrasive access to infected tissues and high abundance of inflammatory mediators that may mask mechanistic details. In diseases such as influenza and smallpox where the primary cause of mortality results from complications in the lung, the characterization of lung fluid offers a unique opportunity to study host-pathogen interactions with minimal effect on infected animals. This investigation characterizes the global proteome response in the pulmonary fluid, bronchoalveolar lavage fluid, of macaques during upper respiratory infection by monkeypox virus (MPXV), a close relative of the causative agent of smallpox, variola virus. These results are compared and contrasted against infections by vaccinia virus (VV), a low pathogenic relative of MPXV, and with extracellular fluid from MPXV-infected HeLa cells. To identify changes in the pulmonary protein compartment, macaque lung fluid was sampled twice prior to infection, serving as base line, and up to six times following intrabronchial infection with either MPXV or VV. Increased expression of inflammatory proteins was observed in response to both viruses. Although the increased expression resolved for a subset of proteins, such as C-reactive protein, S100A8, and S100A9, high expression levels persisted for other proteins, including vitamin D-binding protein and fibrinogen γ. Structural and metabolic proteins were substantially decreased in lung fluid exclusively during MPXV and not VV infection. Decreases in structural and metabolic proteins were similarly observed in the extracellular fluid of MPXV-infected HeLa cells. Results from this study suggest that the host inflammatory response may not be the only facilitator of viral pathogenesis, but rather maintaining pulmonary structural integrity could be a key factor influencing disease progression and mortality.


Asunto(s)
Líquidos Corporales/metabolismo , Pulmón/metabolismo , Mpox/metabolismo , Proteoma , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Animales , Células HeLa , Humanos , Macaca , Datos de Secuencia Molecular , Mpox/inmunología , Reacción en Cadena de la Polimerasa , Espectrometría de Masas en Tándem , Proteínas Virales/química
11.
Vaccine ; 28 Suppl 2: B78-84, 2010 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-20510749

RESUMEN

A high level of genetic and physiological homology with humans has rendered non-human primates (NHP) an essential animal model for biomedical research. As such NHP offer a unique opportunity to study host-pathogen interactions in a species that closely mimics human biology but can yet be maintained under tight laboratory conditions. Indeed, studies using NHP have been critical to our understanding of pathogenesis as well as the development of vaccines and therapeutics. This further facilitated by the fact that NHPs are susceptible to a variety of pathogens that bear significant homology to human pathogens. Unfortunately, these same viruses pose a potential health issue to humans. In this review we discuss the simian herpesviruses and their potential to cause disease in researchers that come into close contact with them.


Asunto(s)
Herpesvirus Cercopitecino 1/patogenicidad , Zoonosis/virología , Animales , Herpesvirus Cercopitecino 1/genética , Humanos , Primates/virología , Riesgo , Zoonosis/transmisión
12.
Cytokine ; 43(2): 220-8, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18639466

RESUMEN

Monkeypox virus (MPV) is an orthopoxvirus with considerable homology to variola major, the etiologic agent of smallpox. Although smallpox was eradicated in 1976, the outbreak of MPV in the U.S. highlights the health hazards associated with zoonotic infections. Like other orthopoxviruses, MPV encodes a secreted chemokine binding protein, vCCI that is abundantly expressed and secreted from MPV infected cells. EMSA data shows vCCI efficiently binds rhesus MIP-1alpha (rhMIP-1alpha) at near one to one stoichiometry. In vitro chemotaxis experiments demonstrate that vCCI completely inhibits rhMIP-1alpha mediated chemotaxis, while in vivo recruitment assays in rhesus macaques using chemokine-saturated implants show a decrease in the number of CD14(+) cells responding to rhMIP-1alpha when vCCI is present, suggesting vCCI is effectively inhibiting chemokine function both in vitro and in vivo. More importantly, we demonstrate that vCCI can diminish the severity of the acute phase and completely inhibit the relapsing phase of experimental allergic encephalomyelitis (EAE) disease. These data represent the first in vitro and in vivo characterization of vCCI emphasizing its function as a potent inhibitor of rhMIP-1alpha. Furthermore, the ability of vCCI to inhibit relapsing EAE disease represents a novel therapeutic approach for treating chemokine-mediated diseases.


Asunto(s)
Proteínas Inflamatorias de Macrófagos/antagonistas & inhibidores , Monkeypox virus/metabolismo , Proteínas Virales/farmacología , Secuencia de Aminoácidos , Animales , Línea Celular , Encefalomielitis Autoinmune Experimental/metabolismo , Humanos , Receptores de Lipopolisacáridos/metabolismo , Macaca mulatta , Proteínas Inflamatorias de Macrófagos/metabolismo , Datos de Secuencia Molecular , Unión Proteica , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Proteínas Virales/química , Proteínas Virales/metabolismo
13.
J Proteome Res ; 7(3): 960-8, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18205298

RESUMEN

Orthopoxviruses are among the largest and most complex of the animal viruses. In response to the recent emergence of monkeypox in Africa and the threat of smallpox bioterrorism, two orthopoxviruses with different pathogenic potentials, human monkeypox virus and vaccinia virus, were proteomically compared with the goal of identifying proteins required for pathogenesis. Orthopoxviruses were grown in HeLa cells to two different viral forms (intracellular mature virus and extracellular enveloped virus), purified by sucrose gradient ultracentrifugation, denatured using RapiGest surfactant, and digested with trypsin. Unfractionated samples and strong cation exchange HPLC fractions were analyzed by high-resolution reversed-phase nano-LC-MS/MS, and analyses of the MS/MS spectra using SEQUEST and X! Tandem resulted in the confident identification of hundreds of monkeypox, vaccinia, and copurified host-cell proteins. The unfractionated samples were additionally analyzed by LC-MS using an LTQ-Orbitrap, and the accurate mass and elution time tag approach was used to perform quantitative comparisons. Possible pathophysiological roles of differentially abundant Orthopoxvirus proteins are discussed. Data, processed results, and protocols are available at http://www.proteomicsresource.org/.


Asunto(s)
Monkeypox virus/química , Proteómica , Virus Vaccinia/química , Virión/química , Cromatografía Líquida de Alta Presión , Cromatografía Liquida/métodos , Electroforesis en Gel de Poliacrilamida , Espectrometría de Masas en Tándem/métodos
14.
J Biomol Tech ; 19(5): 285-95, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19183792

RESUMEN

Mass spectrometry-based proteomics is a powerful analytical tool for investigating pathogens and their interactions within a host. The sensitivity of such analyses provides broad proteome characterization, but the sample-handling procedures must first be optimized to ensure compatibility with the technique and to maximize the dynamic range of detection. The decision-making process for determining optimal growth conditions, preparation methods, sample analysis methods, and data analysis techniques in our laboratory is discussed herein with consideration of the balance in sensitivity, specificity, and biomass losses during analysis of host-pathogen systems.


Asunto(s)
Interacciones Huésped-Patógeno , Proteoma/aislamiento & purificación , Proteómica/métodos , Anaplasma phagocytophilum/química , Anaplasma phagocytophilum/patogenicidad , Animales , Biotecnología , Cromatografía Líquida de Alta Presión , Ehrlichia chaffeensis/química , Ehrlichia chaffeensis/patogenicidad , Células HeLa , Humanos , Espectrometría de Masas/métodos , Espectrometría de Masas/estadística & datos numéricos , Monkeypox virus/química , Monkeypox virus/fisiología , Proteómica/estadística & datos numéricos , Salmonella/química , Salmonella/patogenicidad , Sensibilidad y Especificidad , Biología de Sistemas/métodos , Biología de Sistemas/estadística & datos numéricos , Espectrometría de Masas en Tándem , Virus Vaccinia/química , Virus Vaccinia/patogenicidad
15.
J Virol ; 81(6): 2957-69, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17215283

RESUMEN

Rhesus rhadinovirus (RRV) is closely related to Kaposi's sarcoma-associated herpesvirus (KSHV)/human herpesvirus 8 (HHV-8) and causes KSHV-like diseases in immunocompromised rhesus macaques (RM) that resemble KSHV-associated diseases including multicentric Castleman's disease and non-Hodgkin's lymphoma. RRV retains a majority of open reading frames (ORFs) postulated to be involved in the pathogenesis of KSHV and is the closest available animal model to KSHV infection in humans. Here we describe the generation of a recombinant clone of RRV strain 17577 (RRV(17577)) utilizing bacterial artificial chromosome (BAC) technology. Characterization of the RRV BAC demonstrated that it is a pathogenic molecular clone of RRV(17577), producing virus that behaves like wild-type RRV both in vitro and in vivo. Specifically, BAC-derived RRV displays wild-type growth properties in vitro and readily infects simian immunodeficiency virus-infected RM, inducing B cell hyperplasia, persistent lymphadenopathy, and persistent infection in these animals. This RRV BAC will allow for rapid genetic manipulation of the RRV genome, facilitating the creation of recombinant versions of RRV that harbor specific alterations and/or deletions of viral ORFs. This system will provide insights into the roles of specific RRV genes in various aspects of the viral life cycle and the RRV-associated pathogenesis in vivo in an RM model of infection. Furthermore, the generation of chimeric versions of RRV containing KSHV genes will allow analysis of the function and contributions of KSHV genes to viral pathogenesis by using a relevant primate model system.


Asunto(s)
Cromosomas Artificiales Bacterianos , Herpesvirus Humano 8/genética , Macaca mulatta/virología , Rhadinovirus/genética , Sarcoma de Kaposi/etiología , Animales , Células Cultivadas , ADN Viral/análisis , Fibroblastos/virología , Herpesvirus Humano 8/aislamiento & purificación , Humanos , Hibridación de Ácido Nucleico , Sistemas de Lectura Abierta , Reacción en Cadena de la Polimerasa , Rhadinovirus/química , Sarcoma de Kaposi/virología , Análisis de Secuencia de ADN
16.
J Virol ; 80(6): 3098-103, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16501121

RESUMEN

A viral CD200 homologue (vCD200) encoded by open reading frame R15 of rhesus rhadinovirus (RRV), a gammaherpesvirus closely related to human herpesvirus 8 (HHV-8), is described here. RRV vCD200 shares 30% and 28% amino acid identity with human CD200 (huCD200) and HHV-8 vCD200, respectively. In vitro analysis indicated that an Fc fusion (vCD200-Fc) is expressed as a glycoprotein with a core molecular mass of 53 kDa. Utilizing monoclonal antibodies raised against vCD200-Fc, vCD200 expression was detected on the surfaces of and within supernatants from infected fibroblasts. Furthermore, in vitro assays demonstrated that vCD200-Fc treatment of monocyte-derived macrophages reduces tumor necrosis factor transcript and protein levels, implying that RRV encodes a functional vCD200.


Asunto(s)
Antígenos CD/metabolismo , Macaca mulatta/virología , Rhadinovirus/genética , Rhadinovirus/patogenicidad , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Animales , Antígenos CD/química , Antígenos CD/genética , Células CHO , Cricetinae , Fibroblastos/virología , Humanos , Macrófagos/virología , Datos de Secuencia Molecular , Rhadinovirus/metabolismo , Homología de Secuencia de Aminoácido , Proteínas Virales/química , Proteínas Virales/genética
17.
J Virol ; 79(6): 3878-82, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15731284

RESUMEN

Rhesus macaque rhadinovirus (RRV) is the rhesus macaque homologue of human herpesvirus 8 (HHV-8). Here we examine expression of RRV R15 and ORF74, homologues of K14 and ORF74 of HHV-8, respectively. As in HHV-8, transcripts encoding RRV R15 and ORF74 are bicistronic. However, unlike what has been suggested for HHV-8, RRV R15- and ORF74-encoding transcripts are expressed late during lytic infection and undergo unique splicing events that result in the production of transcripts capable of encoding vGPCR, as well as membrane-associated and secreted forms of vCD200. The alternative splicing for vCD200 has implications for viral pathogenesis.


Asunto(s)
Sistemas de Lectura Abierta , Precursores del ARN/metabolismo , Empalme del ARN , ARN Viral/metabolismo , Receptores Acoplados a Proteínas G/biosíntesis , Rhadinovirus/genética , Proteínas Virales/biosíntesis , Northern Blotting , Genes Virales , Receptores Acoplados a Proteínas G/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Virales/genética
18.
J Virol ; 77(3): 1738-46, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12525607

RESUMEN

Rhesus rhadinovirus (RRV) is a gamma-2 herpesvirus and is the rhesus macaque homologue of human herpesvirus 8 (HHV-8), also known as Kaposi's sarcoma-associated herpesvirus. DNA sequence analysis of RRV indicates that it shares numerous open reading frames (ORFs) with HHV-8, including one (ORF74) encoding a seven-transmembrane-spanning G protein-coupled receptor (GPCR) with similarity to cellular chemokine receptors. Examination of the predicted amino acid sequence of RRV ORF74 reveals that it encodes a seven-transmembrane-spanning GPCR sharing 40.8% amino acid sequence identity with HHV-8 ORF74 and 24.1% amino acid sequence identity with rhesus macaque CXCR2. In addition, immunofluorescence studies indicate that an epitope-tagged version of RRV ORF74 is expressed on the surfaces of transfected cells, suggesting that this protein is in fact a membrane receptor. In in vitro cell culture assays, RRV ORF74 possesses transforming potential, as NIH 3T3 clones stably expressing the receptor demonstrate an increased ability to grow in soft agarose and to induce tumor formation in nude mice. Further analysis of RRV ORF74 indicates that expression of the receptor in NIH 3T3 cells causes an increased secretion of vascular endothelial growth factor and activation of the ERK1/2 (p44/42) mitogen-activated protein kinase signaling pathway. The results of these studies suggest that RRV ORF74 encodes a GPCR with properties similar to those of its homologue in HHV-8 and that this gene may play a role in RRV-associated pathogenesis.


Asunto(s)
Proteínas de Unión al GTP/fisiología , Macaca mulatta/virología , Sistemas de Lectura Abierta , Receptores de Superficie Celular/química , Receptores de Quimiocina/química , Rhadinovirus/genética , Proteínas Virales/química , Células 3T3 , Secuencia de Aminoácidos , Animales , Transformación Celular Neoplásica , Factores de Crecimiento Endotelial/biosíntesis , Activación Enzimática , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Linfocinas/biosíntesis , Ratones , Ratones Desnudos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Receptores de Interleucina-8B/química , Rhadinovirus/química , Alineación de Secuencia , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...