Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
PLoS One ; 9(3): e91359, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24614587

RESUMEN

Kaposi's sarcoma herpesvirus (KSHV) is a gamma-2 herpesvirus present in all cases of Kaposi's sarcoma, primary effusion lymphoma (PEL), and some cases of multicentric Castleman's disease. Viral FLICE inhibitory protein (vFLIP) is a latently expressed gene that has been shown to be essential for survival of latently infected PEL cells by activating the NFκB pathway. Inhibitors of either vFLIP expression or the NFĸB pathway result in enhanced lytic reactivation and apoptosis. We have observed a decrease in vFLIP protein levels and of NFκB activation in the presence of the KSHV lytic switch protein RTA. Whereas vFLIP alone induced expression of the NFĸB responsive genes ICAM1 and TNFα, inclusion of RTA decreased vFLIP induced ICAM1 and TNFα expression in both co-transfected 293T cells and in doxycycline induced TREx BCBL1 cells. RTA expression resulted in proteasome dependent destabilization of vFLIP. Neither RTA ubiquitin E3 ligase domain mutants nor a dominant-negative RAUL mutant abrogated this effect, while RTA truncation mutants did, suggesting that RTA recruits a novel cellular ubiquitin E3 ligase to target vFLIP for proteasomal degradation, allowing for inhibition of NFĸB responsive gene expression early during lytic reactivation.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Regulación de la Expresión Génica , Herpesvirus Humano 8/metabolismo , FN-kappa B/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Transactivadores/metabolismo , Activación Viral/genética , Animales , Chlorocebus aethiops , Células HEK293 , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Proteolisis , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Regulación hacia Arriba , Células Vero
2.
Cell Host Microbe ; 10(4): 390-400, 2011 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-22018239

RESUMEN

Herpesviruses, which are major human pathogens, establish life-long persistent infections. Although the α, ß, and γ herpesviruses infect different tissues and cause distinct diseases, they each encode a conserved serine/threonine kinase that is critical for virus replication and spread. The extent of substrate conservation and the key common cell-signaling pathways targeted by these kinases are unknown. Using a human protein microarray high-throughput approach, we identify shared substrates of the conserved kinases from herpes simplex virus, human cytomegalovirus, Epstein-Barr virus (EBV), and Kaposi's sarcoma-associated herpesvirus. DNA damage response (DDR) proteins were statistically enriched, and the histone acetyltransferase TIP60, an upstream regulator of the DDR pathway, was required for efficient herpesvirus replication. During EBV replication, TIP60 activation by the BGLF4 kinase triggers EBV-induced DDR and also mediates induction of viral lytic gene expression. Identification of key cellular targets of the conserved herpesvirus kinases will facilitate the development of broadly effective antiviral strategies.


Asunto(s)
Daño del ADN , Enzimas Reparadoras del ADN/metabolismo , Herpesviridae/enzimología , Histona Acetiltransferasas/metabolismo , Interacciones Huésped-Patógeno , Proteínas Serina-Treonina Quinasas/metabolismo , Replicación Viral , Secuencia Conservada , Herpesviridae/fisiología , Humanos , Lisina Acetiltransferasa 5 , Análisis por Micromatrices , Modelos Biológicos , Análisis por Matrices de Proteínas
3.
J Infect Dis ; 204(11): 1683-91, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21990424

RESUMEN

BACKGROUND: Epstein-Barr virus (EBV) is a ubiquitous herpesvirus, and Kaposi's sarcoma-associated herpesvirus (KSHV) has a restricted seroprevalence. Both viruses are associated with malignancies that have an increased frequency in individuals who are coinfected with human immunodeficiency virus type 1 (HIV-1). METHODS: To obtain an overview of humoral immune responses to these viruses, we generated a protein array that displayed 174 EBV and KSHV polypeptides purified from yeast. Antibody responses to EBV and KSHV were examined in plasma from healthy volunteers and patients with B cell lymphoma or with AIDS-related Kaposi's sarcoma or lymphoma. RESULTS: In addition to the commonly studied antigens, IgG responses were frequently detected to the tegument proteins KSHV ORF38 and EBV BBRF and BGLF2 and BNRF1 and to the EBV early lytic proteins BRRF1 and BORF2. The EBV vIL-10 protein was particularly well recognized by plasma IgA. The most intense IgG responses to EBV antigens occurred in HIV-1-positive patients. No clear correlation was observed between viral DNA load in plasma and antibody profile. CONCLUSIONS: The protein array provided a sensitive platform for global screening; identified new, frequently recognized viral antigens; and revealed a broader humoral response to EBV compared with KSHV in the same patients.


Asunto(s)
Antígenos Virales/sangre , Herpesvirus Humano 4/inmunología , Herpesvirus Humano 8/inmunología , Inmunidad Humoral , Análisis por Matrices de Proteínas/métodos , Seronegatividad para VIH/inmunología , Seropositividad para VIH/inmunología , VIH-1/inmunología , Humanos , Inmunoglobulina A/inmunología , Linfoma Relacionado con SIDA/sangre , Linfoma Relacionado con SIDA/inmunología , Linfoma Relacionado con SIDA/virología , Linfoma de Células B/sangre , Linfoma de Células B/inmunología , Linfoma de Células B/virología , Sarcoma de Kaposi/sangre , Sarcoma de Kaposi/inmunología , Sarcoma de Kaposi/virología , Carga Viral/inmunología
4.
Am J Pathol ; 179(3): 1443-54, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21741351

RESUMEN

Kaposi's sarcoma is an angioproliferative tumor caused by Kaposi's sarcoma-associated herpesvirus (KSHV) infection of vascular endothelial cells. Fibulins, proteins that associate with extracellular matrix (ECM) proteins, may have both tumor-suppressive and oncogenic activities. We found that the expression of fibulin-2 protein and mRNA were decreased 50-fold and 26-fold, respectively, in 10-day KSHV-infected dermal microvascular endothelial cells (DMVEC). Using quantitative RT-PCR, we found a fivefold and 25-fold decrease of fibulin-2 extracellular matrix binding partners, fibronectin and tropoelastin, respectively. Time-course transcriptional analyses over 10 days showed that in addition to that of fibulin-2, expression of fibulins 3 and 5 was decreased in KSHV-infected DMVEC, fibulins 1C/1D were increased, and fibulins 4, 6, and 7 were unchanged. KSHV latency-associated nuclear antigen (LANA) transcription levels rose consistently over the same period. Addition of recombinant fibulin-3 or -5 for 48 hours to 10-day KSHV-infected cells caused a suppression of KSHV-induced vascular endothelial growth factor (VEGF) protein and mRNA levels. Recombinant fibulin-3 also significantly reduced VEGF receptor 3 expression. In pleural effusion lymphoma cell lines that express variable levels of KSHV lytic replication, we observed no detectable fibulin-2 or -5 expression. Finally, fibulin-2 expression was decreased in tissue microarrays from KSHV-infected, LANA-positive patient cells as compared to that in patient nontumor controls. Understanding the interactions between KSHV and the fibulins may lead to the development of novel therapies for treatment of Kaposi's sarcoma.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Transformación Celular Neoplásica/patología , Proteínas de la Matriz Extracelular/metabolismo , Herpesvirus Humano 8/fisiología , Sarcoma de Kaposi/virología , Neoplasias Cutáneas/virología , Línea Celular Tumoral , Regulación hacia Abajo , Endotelio Vascular/virología , Proteínas de la Matriz Extracelular/farmacología , Fibronectinas/metabolismo , Humanos , Proteínas Recombinantes , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/patología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Tropoelastina/metabolismo , Regulación hacia Arriba , Carga Viral
5.
Vet Microbiol ; 147(1-2): 28-41, 2011 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-20579821

RESUMEN

Systemic infections with elephant endotheliotropic herpesviruses (EEHV) cause a rapid onset acute hemorrhagic disease with an 85% mortality rate. More than 60 cases have been confirmed worldwide occurring predominantly in juvenile Asian elephants. Originally, three virus types EEHV1A, EEHV1B and EEHV2 were identified, all members of the Proboscivirus genus within the Betaherpesvirinae. However, four elephant gammaherpesviruses (EGHV) have also been found by DNA PCR approaches in eye and genital secretions of asymptomatic animals, and two more versions of the probosciviruses, EEHV3 and EEHV4, were recently detected in acute hemorrhagic disease cases. To ask whether even more species of elephant herpesviruses may exist, we have developed several new diagnostic DNA PCR assays using multiple round primers in the DNA POL region. These have been used routinely for nearly three years to screen samples submitted to the Elephant Herpesvirus Laboratory for diagnosis of possible cases of EEHV disease in blood and necropsy tissue, as well as in biopsies of other suspicious lesions or growths. Several more cases of EEHV1-associated hemorrhagic disease were confirmed, but in addition, we describe here eleven examples of other known and novel herpesviruses detected and evaluated with these reagents. They include the prototypes of four new elephant herpesviruses, two more within the proboscivirus group EEHV5 and EEHV6, plus two more gammaherpesviruses EGHV3B and EGHV5. We also report initial semi-quantitative PCR assays demonstrating very high viral loads in the blood of the EEHV3 and EEHV4-associated hemorrhagic disease cases.


Asunto(s)
Betaherpesvirinae , Elefantes/virología , Gammaherpesvirinae , Infecciones por Herpesviridae/veterinaria , Filogenia , Secuencia de Aminoácidos , Animales , Betaherpesvirinae/clasificación , Betaherpesvirinae/genética , Betaherpesvirinae/aislamiento & purificación , ADN Polimerasa Dirigida por ADN/química , ADN Polimerasa Dirigida por ADN/genética , Femenino , Gammaherpesvirinae/clasificación , Gammaherpesvirinae/genética , Gammaherpesvirinae/aislamiento & purificación , Infecciones por Herpesviridae/virología , Masculino , Datos de Secuencia Molecular , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Especificidad de la Especie , Carga Viral
6.
Immunity ; 33(6): 863-77, 2010 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-21167755

RESUMEN

In the course of combating infectious agents, type I interferon (IFN) needs a timely downregulation mechanism to avoid detrimental overreaction. Here we showed a mechanism for restraining type I IFN responses, which relied on a HECT domain ubiquitin (Ub) E3 ligase, RAUL. RAUL limited type I IFN production by directly catalyzing lysine 48-linked polyubiquitination of both interferon regulatory factor 7 (IRF7) and IRF3 followed by proteasome-dependent degradation. Suppression of RAUL by dominant-negative RAUL or siRNA augmented both basal and virus-induced production of type I IFN, which resulted in reduced viral replication. The Kaposi's sarcoma-associated herpes virus immediate-early lytic cycle trigger protein RTA recruited this mechanism to augment its countermeasures against the host antiviral response. These results unveil a previously unrecognized "brake mechanism" for type I IFN that maintains proper low amounts of type I IFN under physiological conditions and restrains its magnitude when the antiviral response intensifies.


Asunto(s)
Fibroblastos/metabolismo , Infecciones por Herpesviridae/inmunología , Herpesvirus Humano 8/fisiología , Interferón Tipo I/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular , Regulación hacia Abajo/genética , Fibroblastos/inmunología , Fibroblastos/patología , Fibroblastos/virología , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/metabolismo , Herpesvirus Humano 8/patogenicidad , Humanos , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Ratones , Proteínas Mutantes/genética , Complejo de la Endopetidasa Proteasomal , ARN Interferente Pequeño/genética , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología , Ubiquitinación/genética , Replicación Viral/genética
7.
J Gen Virol ; 91(Pt 3): 616-21, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19923265

RESUMEN

The immediate-early 1 (IE1) and IE2 proteins encoded by the major immediate-early (MIE) transcription unit of cytomegaloviruses are thought to play key roles in the switch between latent- and lytic-cycle infection. Whilst IE2 is essential for triggering the lytic cycle, the exact roles of IE1 have not been resolved. An MIE-exon 4-deleted rat cytomegalovirus (DeltaIE1) failed to synthesize the IE1 protein and did not disperse promyelocytic leukaemia bodies early post-infection, but was still capable of normal replication in fibroblast cell culture. However, DeltaIE1 had a diminished ability to infect salivary glands persistently in vivo and to reactivate from spleen explant cultures ex vivo. Quantification of viral genomes in spleens of infected animals revealed a reduced amount of DeltaIE1 virus produced during acute infection, suggesting a role for IE1 as a regulator in establishing a chronic or persistent infection, rather than in influencing the latency or reactivation processes more directly.


Asunto(s)
ADN Viral/genética , Proteínas Inmediatas-Precoces/genética , Muromegalovirus/fisiología , Eliminación de Secuencia , Transactivadores/genética , Latencia del Virus , Replicación Viral , Animales , Células Cultivadas , Fibroblastos/virología , Infecciones por Herpesviridae/virología , Técnicas In Vitro , Muromegalovirus/genética , Ratas , Glándulas Salivales/virología , Bazo/virología , Virulencia , Activación Viral
8.
Glycobiology ; 20(5): 521-32, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20040515

RESUMEN

Galectins are a family of proteins that share an affinity for beta-galactoside containing glycoconjugates. In prostate, ovarian and breast cancer, downregulation of galectin-3 is associated with malignancy and tumor progression. Kaposi's sarcoma (KS) is characterized as an angioproliferative tumor of vascular endothelial cells and produces rare B cell lymphoproliferative diseases in the form of primary effusion lymphomas and some forms of multicentric Castleman's disease. Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent of KS. We found reduced levels of galectin-3 expression in a significant fraction of latency-associated nuclear antigen (LANA)-positive spindle cell regions in human archival KS tissue and as measured in KS tissue microarrays. Here we demonstrate that galectin-3 protein expression is downregulated 10-fold in 10-day KSHV-infected dermal microvascular endothelial cells (DMVEC) accompanied by downregulation of message. There is loss of galectin-3 staining in KSHV-infected DMVEC by dual labeled immunohistochemistry in LANA-positive spindle cells. We observed a consistent downregulation of galectin-3 by time-course transcriptional analysis. Of the galectins assayed, only galectin-1 was also downregulated in KSHV-infected DMVEC. We examined 86 KS tumors; 19 were LANA positive (22%) and 67 LANA negative (78%). All 86 tumors were found to be galectin-3 positive; 11 of 19 showed reduced expression of galectin-3 in LANA-positive spindle cell regions. Our data suggest that KSHV vFLIP and LANA are the viral genes targeting galectin-3 downregulation. The contribution of host factors to the pathogenesis of KS is essential for early detection and development of innovative therapies for treatment.


Asunto(s)
Regulación hacia Abajo , Galectina 3/metabolismo , Herpesvirus Humano 8/fisiología , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virología , Antígenos Virales/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/virología , Galectina 3/biosíntesis , Galectina 3/genética , Células HeLa , Humanos , Inmunohistoquímica , Microcirculación , Proteínas Nucleares/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Matrices Tisulares , Células Tumorales Cultivadas
9.
J Clin Oncol ; 27(15): 2496-502, 2009 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-19349542

RESUMEN

PURPOSE: To determine the relative frequency with which Kaposi's sarcoma-associated herpesvirus/HHV-8 (KSHV) DNA is detected in peripheral-blood mononuclear cells (PBMCs) and in plasma of patients with AIDS-KS and AIDS-associated non-Hodgkin's lymphoma (NHL; AIDS-NHL); to determine whether the presence of viral DNA in plasma reflects lysis of tumor cells or reflects the presence of viremia (ie, virion-encapsidated DNA); and to determine the effect of lymphoma therapy on KSHV DNA. PATIENTS AND METHODS: Samples were obtained from patients enrolled in AIDS Malignancy Consortium clinical trials and from healthy donors. Real time PCR was used to quantify KSHV DNA in peripheral blood mononuclear cells (PBMC) and plasma. DNase digestion and fragment size determination studies were used to characterize the DNA detected. RESULTS: In patients with AIDS-KS, KSHV DNA was detected in PBMC (54%) and in plasma (62%). In patients with AIDS-NHL, KSHV DNA was detected in PBMC (19%) and in plasma (22%). Median copy numbers also differed. KSHV DNA in plasma appeared to be encapsidated. In six patients with AIDS-NHL who were treated with chemotherapy (with or without rituximab), KSHV copy number declined in PBMC and in plasma. CONCLUSION: KSHV DNA is sometimes detected in PBMC or in plasma of patients with AIDS-NHL without KS. Among patients with KSHV DNA detected in PBMC or in plasma, copy number does not distinguish between patients with AIDS-NHL and AIDS-KS. KSHV DNA in plasma likely reflects viremia and not simply lysis of tumor or other KSHV-infected cells. KSHV DNA copy number in PBMC and in plasma declined with lymphoma-directed cytotoxic chemotherapy in each of the six patients studied.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/sangre , Antineoplásicos/uso terapéutico , ADN Viral/efectos de los fármacos , Herpesvirus Humano 8/aislamiento & purificación , Linfoma Relacionado con SIDA/sangre , Linfoma no Hodgkin/sangre , Síndrome de Inmunodeficiencia Adquirida/complicaciones , Ensayos Clínicos como Asunto , Herpesvirus Humano 8/genética , Humanos , Leucocitos Mononucleares/virología , Linfoma Relacionado con SIDA/tratamiento farmacológico , Linfoma Relacionado con SIDA/virología , Linfoma no Hodgkin/tratamiento farmacológico , Linfoma no Hodgkin/virología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcoma de Kaposi/sangre , Sarcoma de Kaposi/virología
10.
J Virol ; 83(10): 5219-31, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19244323

RESUMEN

A conserved family of herpesvirus protein kinases plays a crucial role in herpesvirus DNA replication and virion production. However, despite the fact that these kinases are potential therapeutic targets, no systematic studies have been performed to identify their substrates. We generated an Epstein-Barr virus (EBV) protein array to evaluate the targets of the EBV protein kinase BGLF4. Multiple proteins involved in EBV lytic DNA replication and virion assembly were identified as previously unrecognized substrates for BGLF4, illustrating the broad role played by this protein kinase. Approximately half of the BGLF4 targets were also in vitro substrates for the cellular kinase CDK1/cyclin B. Unexpectedly, EBNA1 was identified as a substrate and binding partner of BGLF4. EBNA1 is essential for replication and maintenance of the episomal EBV genome during latency. BGLF4 did not prevent EBNA1 binding to sites in the EBV latency origin of replication, oriP. Rather, we found that BGLF4 was recruited by EBNA1 to oriP in cells transfected with an oriP vector and BGLF4 and in lytically induced EBV-positive Akata cells. In cells transfected with an oriP vector, the presence of BGLF4 led to more rapid loss of the episomal DNA, and this was dependent on BGLF4 kinase activity. Similarly, expression of doxycycline-inducible BGLF4 in Akata cells led to a reduction in episomal EBV genomes. We propose that BGLF4 contributes to effective EBV lytic cycle progression, not only through phosphorylation of EBV lytic DNA replication and virion proteins, but also by interfering with the EBNA1 replication function.


Asunto(s)
Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/metabolismo , Análisis por Matrices de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Virales/metabolismo , Línea Celular Tumoral , Replicación del ADN , ADN Viral/metabolismo , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/fisiología , Humanos , Fosforilación , Especificidad por Sustrato , Replicación Viral
11.
J Virol ; 82(21): 10444-54, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18701593

RESUMEN

The human cytomegalovirus (HCMV) 72-kDa immediate-early 1 (IE1) protein is thought to modulate cellular antiviral functions impacting on promyelocytic leukemia (PML) nuclear bodies and signal transducer and activator of transcription (STAT) signaling. IE1 consists of four distinct regions: an amino-terminal region required for nuclear localization, a large central hydrophobic region responsible for PML targeting and transactivation activity, an acidic domain, and a carboxyl-terminal chromatin tethering domain. We found that the acidic domain of IE1 is required for binding to STAT2. A mutant HCMV encoding IE1(Delta421-475) with the acidic domain deleted was generated. In mutant virus-infected cells, IE1(Delta421-475) failed to bind to STAT2. The growth of mutant virus was only slightly delayed at a high multiplicity of infection (MOI) but was severely impaired at a low MOI with low-level accumulation of viral proteins. When cells were pretreated with beta interferon, the mutant virus showed an additional 1,000-fold reduction in viral growth, even at a high MOI, compared to the wild type. The inhibition of STAT2 loading on the target promoter upon infection was markedly reduced with mutant virus. Furthermore, sumoylation of IE1 at this acidic domain was found to abolish the activity of IE1 to bind to STAT2 and repress the interferon-stimulated genes. Our results provide genetic evidence that IE1 binding to STAT2 requires the 55-amino-acid acidic domain and promotes viral growth by interfering with interferon signaling and demonstrate that this viral activity is negatively regulated by a cellular sumoylation pathway.


Asunto(s)
Citomegalovirus/crecimiento & desarrollo , Proteínas Inmediatas-Precoces/metabolismo , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Factor de Transcripción STAT2/metabolismo , Proteína SUMO-1/metabolismo , Línea Celular , Células Cultivadas , Humanos , Unión Proteica , Eliminación de Secuencia
12.
J Clin Virol ; 40(1): 19-25, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17690010

RESUMEN

BACKGROUND: Small 233-bp or 330-bp DNA fragments of the ORF26 gene of human Kaposi's sarcoma herpesvirus (KSHV) have been used extensively to identify KSHV by PCR in clinical samples; to associate KSHV with novel diseases and to correlate KSHV strain differences with pathogenicity. OBJECTIVES: We evaluated the nature, extent and source of nucleotide sequence variability among a large and diverse set of known KSHV-positive DNA samples. STUDY DESIGN: Direct DNA PCR sequencing was carried out on 136 distinct Kaposi's sarcoma and primary effusion lymphoma-related samples from different geographic locations. RESULTS: The presence of 26 diagnostic nucleotide polymorphisms across an expanded 965-bp PCR locus define eight distinct ORF26E genotypes, three being of Eurasian origin, one from the Pacific Rim, and five from Sub-Saharan Africa. Previous ambiguities between some genotype patterns in the 330-bp locus data are fully resolved. CONCLUSIONS: This analysis provides an expanded database for understanding and evaluating ORF26 polymorphisms. In particular, the eight genotype clusters correlated with specific ethnic and geographic origins of the patients. Furthermore, the very low level of additional sporadic nucleotide variation found permits detection of spurious sequence errors or contamination present in some published data.


Asunto(s)
Infecciones por Herpesviridae/epidemiología , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/aislamiento & purificación , Sistemas de Lectura Abierta , Polimorfismo Genético , Sarcoma de Kaposi/epidemiología , Infecciones Oportunistas Relacionadas con el SIDA/virología , Secuencia de Bases , Análisis por Conglomerados , Demografía , Genoma Viral , Genotipo , Infecciones por Herpesviridae/virología , Humanos , Datos de Secuencia Molecular , Sarcoma de Kaposi/virología
13.
J Clin Virol ; 40(1): 1-8, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17698410

RESUMEN

Ever since the original identification of fragments of KSHV DNA in Kaposi's sarcoma (KS) tissue by Chang et al. in 1994, PCR has been used successfully and extensively to detect the virus in clinical samples from the accepted etiological diseases of KS, PEL and MCD. However, a number of other clinical and epidemiological studies claiming evidence for KSHV in multiple myeloma or sarcoid and more recently in primary pulmonary hypertension, as well as claims about the biological significance of DNA sequence polymorphisms based just on small ORF26 PCR DNA fragments have not been convincing. Here, we evaluate the validity and interpretations of previous results in the context of both the observed rates and global patterns of sequence variability within an extended ORF26 locus, as well as from the perspective of the overall levels of KSHV variability found after sampling multiple loci across the complete KSHV genome. The results cast doubts on most claims for biological significance for these polymorphisms, which instead correlate with viral subtype clustering arising from geographic and ethnic divergence of the ancestral human hosts. In addition, we describe several observations that help to explain likely sources of the often either unexpectedly high or unexpectedly low levels of sporadic variability seen in the PCR DNA sequence data reported in some of those studies.


Asunto(s)
Genoma Viral , Herpesvirus Humano 8/genética , Sarcoma de Kaposi/epidemiología , Sarcoma de Kaposi/virología , Infecciones Oportunistas Relacionadas con el SIDA/epidemiología , Infecciones Oportunistas Relacionadas con el SIDA/virología , Variación Genética , Genotipo , Infecciones por Herpesviridae/epidemiología , Infecciones por Herpesviridae/virología , Humanos , Sistemas de Lectura Abierta , Reacción en Cadena de la Polimerasa , Polimorfismo Genético
14.
J Infect Dis ; 194(4): 464-73, 2006 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16845629

RESUMEN

BACKGROUND: Cytomegalovirus (CMV) is the most common congenital infection in humans. The effect of viral strains on the outcome of congenital CMV is debated. We evaluated whether UL144 polymorphisms in amniotic fluid from CMV-infected Italian women were associated with terminations of pregnancy, subsequent disease in their offspring, or viral load. METHODS: The study was nested within a prenatal CMV program. We sequenced the UL144 gene from 66 amniotic-fluid samples, without knowledge of pregnancy outcome. We performed data analyses on 56 samples for which all information was available. RESULTS: Genotype C was associated with termination of pregnancy (P=.03). Genotype B was associated with fewer terminations of pregnancy (P=.003). A possible association was found between genotype C and symptomatic disease in newborns (odds ratio, 8.81 [95% confidence interval, 0.48-164.02]; P=.05). There was no association between specific genotype and the viral load in amniotic fluid. Symptomatic newborns who had the most common UL144 genotype (B) were more likely to have higher viral loads than were asymptomatic infants (P=.003). CONCLUSIONS: UL144 polymorphisms may be associated with the outcome of congenital CMV infection. Larger studies should be conducted to confirm this association, before genotype analysis can be used, along with other factors, in considering terminations of pregnancy.


Asunto(s)
Infecciones por Citomegalovirus/virología , Citomegalovirus/genética , Transmisión Vertical de Enfermedad Infecciosa , Complicaciones Infecciosas del Embarazo/virología , Líquido Amniótico/virología , Citomegalovirus/patogenicidad , Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/transmisión , Femenino , Feto/virología , Humanos , Italia , Glicoproteínas de Membrana/genética , Polimorfismo Genético , Embarazo , Resultado del Embarazo , Receptores del Factor de Necrosis Tumoral/genética , Receptores Virales/genética , Proteínas Virales/genética
15.
Intervirology ; 49(3): 133-43, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16428889

RESUMEN

OBJECTIVE: The aim of this study was to test the relationship between Kaposi's sarcoma-associated herpesvirus (KSHV) phylogeny and host ethnicity at the within-country scale. METHODS: KSHV genomic DNA samples were isolated from 31 patients across eleven Ugandan ethnic groups. Amino acid sequences of the ORF-K1 gene were used to construct a neighbor-joining phylogenetic tree. RESULTS: A5 and B1 variants predominated with no evidence of distinct ethnic or geographic distribution. A new K1 subtype (F) was identified in a member of the Bantu Gisu tribe and a new subtype B variant (B3) among members of the Bantu Ganda tribe. CONCLUSIONS: The phylogeny may yet be structured by host ethnicity if members of Ugandan groups have convoluted biological origins, even as they identify with single tribes. An alternative possibility is that KSHV subtype evolution may have preceded major diversification of sub-Saharan Africans into ethnicities as we know them today, with ethnic groups beginning their histories already hosting multiple subtypes. A third alternative is that horizontal transmission of multiple KSHV subtypes may have broken up vertical lineages of the virus passed down within Ugandan populations.


Asunto(s)
Transmisión de Enfermedad Infecciosa , Infecciones por Herpesviridae/transmisión , Herpesvirus Humano 8/clasificación , Filogenia , Sarcoma de Kaposi/virología , Proteínas Virales/genética , Secuencia de Aminoácidos , ADN Viral/análisis , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Datos de Secuencia Molecular , Sarcoma de Kaposi/etnología , Alineación de Secuencia , Análisis de Secuencia de Proteína , Uganda/epidemiología , Proteínas Virales/metabolismo
16.
J Virol ; 80(3): 1098-109, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16414987

RESUMEN

The contribution of C/EBP proteins to Epstein-Barr virus (EBV) lytic gene expression and replication in epithelial cells was examined. Nasopharyngeal carcinoma cell lines constitutively expressed C/EBPbeta and had limited C/EBPalpha expression, while the AGS gastric cancer cell line expressed significant levels of both C/EBPalpha and C/EBPbeta. Induction of the lytic cycle in EBV-positive AGS/BX1 cells with phorbol ester and sodium butyrate treatment led to a transient stimulation of C/EBPbeta expression and a prolonged increase in C/EBPalpha expression. In AGS/BX1 cells, endogenous C/EBPalpha and C/EBPbeta proteins were detected associated with the ZTA and oriLyt promoters but not the RTA promoter. Electrophoretic mobility shift assays confirmed binding of C/EBP proteins to multiple sites in the ZTA and oriLyt promoters. The response of these promoters in reporter assays to transfected C/EBPalpha and C/EBPbeta proteins was consistent with the promoter binding assays and emphasized the relative importance of C/EBPs for activation of the ZTA promoter. Mutation of the oriLyt promoter proximal C/EBP site had little effect on ZTA activation of the promoter in a reporter assay. However, this mutation impaired oriLyt DNA replication, suggesting a separate replication-specific contribution for C/EBP proteins. Finally, the overall importance of C/EBP proteins for lytic gene expression was demonstrated using CHOP10 to antagonize C/EBP DNA binding activity. Introduction of CHOP10 significantly impaired induction of the ZTA, RTA, and BMRF1 proteins in chemically treated AGS/BX1 cells. Thus, C/EBPbeta and C/EBPalpha expression are associated with lytic induction in AGS cells, and expression of C/EBP proteins in epithelial cells may contribute to the tendency of these cells to exhibit constitutive low-level ZTA promoter activity.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/fisiología , Antígenos Virales/biosíntesis , Antígenos Virales/genética , Secuencia de Bases , Proteínas Potenciadoras de Unión a CCAAT/genética , Línea Celular Tumoral , ADN Viral/genética , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Células Epiteliales/metabolismo , Células Epiteliales/virología , Expresión Génica , Genes Virales , Células HeLa , Humanos , Proteínas Inmediatas-Precoces/biosíntesis , Proteínas Inmediatas-Precoces/genética , Regiones Promotoras Genéticas , Transactivadores/biosíntesis , Transactivadores/genética , Proteínas Virales/biosíntesis , Proteínas Virales/genética , Replicación Viral
17.
Immunity ; 22(1): 59-70, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15664159

RESUMEN

Many viruses encode proteins that counteract the development of the interferon (IFN)-mediated antiviral state. Here, we report that interferon regulatory factor 7 (IRF7), a key mediator of type I IFN induction, is targeted for degradation by binding to the RTA immediate-early nuclear transcription factor encoded by Kaposi's sarcoma-associated herpesvirus (KSHV or HHV8). Cotransfection with RTA blocked IRF7-mediated IFNalpha and IFNbeta mRNA production and promoted the ubiquitination and degradation of IRF7 protein in a proteasome-dependent fashion. Addition of RTA also promoted polyubiquitination of IRF7 in an in vitro cell free assay, demonstrating that RTA itself acts as a ubiquitin E3 ligase. RTA also autoregulated its own polyubiquitination and stability, and both activities were abolished by point mutations in a Cys plus His-rich N-terminal domain. Therefore, manipulation of the stability and function of IRF7 by the KSHV RTA transcription factor provides an unexpected regulatory strategy for circumventing the innate immune defence system.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación Viral de la Expresión Génica , Transactivadores/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Vectores Genéticos , Células HeLa , Herpesvirus Humano 8/genética , Humanos , Factor 7 Regulador del Interferón , Mutagénesis Sitio-Dirigida , Fosforilación , Plásmidos , Regiones Promotoras Genéticas , Unión Proteica , Especificidad por Sustrato , Transactivadores/genética , Transfección , Células Tumorales Cultivadas , Activación Viral
18.
J Gen Virol ; 85(Pt 8): 2155-2166, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15269354

RESUMEN

Kaposi's sarcoma (KS)-associated herpesvirus (KSHV) proteins ORF57 (also known as MTA) and ORF50 (also known as RTA) act post-transcriptionally and transcriptionally to regulate viral lytic gene expression and synergistically activate certain early and late KSHV promoters. When ORF57 and ORF50 were co-expressed, they co-operatively stimulated expression from the promoter of the immediate-early ORF50 gene itself. Co-immunoprecipitations with extracts of KSHV-infected cells showed that ORF57 and ORF50 proteins were present in the same complex. Using the pull-down assay with extracts of KSHV-infected cells, ORF50 protein was shown to interact with a glutathione S-transferase-ORF57 fusion protein. A chromatin immunoprecipitation assay showed that ORF50 promoter sequences were preferentially associated with immunoprecipitated chromatin using both anti-ORF50 and anti-ORF57 antibodies consistent with both an in vivo physical association between ORF57 and ORF50 and a potential role for ORF57 at the transcriptional level. This is the first demonstration of an interaction between these two lytic regulatory proteins in a gammaherpesvirus. Expression of ORF50 protein is sufficient to induce lytic replication in latently infected cells and may determine viral host range, spread and KS pathogenesis in vivo. A new insight into the co-ordinated activities of these two key regulatory proteins is provided in which upregulation of the ORF50 promoter with augmentation of ORF50 activity by ORF57 protein, and vice versa, would facilitate the cascade of lytic viral gene expression, thereby breaking latency. A functional and physical interaction between these two gammaherpesvirus regulatory protein counterparts could be a general feature of the herpesviruses.


Asunto(s)
Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/genética , Proteínas Inmediatas-Precoces/fisiología , Transactivadores/fisiología , Proteínas Virales/fisiología , Línea Celular , Humanos , Pruebas de Precipitina , Regiones Promotoras Genéticas , Acetato de Tetradecanoilforbol/farmacología , Transcripción Genética
19.
J Virol ; 78(12): 6527-42, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15163746

RESUMEN

In one of the earliest events in human cytomegalovirus (HCMV)-infected cells, the major immediate-early (IE) protein IE1 initially targets to and then disrupts the nuclear structures known as PML oncogenic domains (PODs) or nuclear domain 10. Recent studies have suggested that modification of PML by SUMO is essential to form PODs and that IE1 both binds to PML and may disrupt PODs by preventing or removing SUMO adducts on PML. In this study, we showed that in contrast to herpes simplex virus type 1 (HSV-1) IE110 (ICP0), the loss of sumoylated forms of PML by cotransfected IE1 was resistant to the proteasome inhibitor MG132 and that IE1 did not reduce the level of unmodified PML. Reduced sumoylation of PML was also observed in U373 cells after infection with wild-type HCMV and proved to require IE1 protein expression. Mutational analysis revealed that the central hydrophobic domain of IE1, including Leu174, is required for both PML binding and loss of PML sumoylation and confirmed that all IE1 mutants tested that were deficient in these functions also failed both to target to PODs and to disrupt PODs. These same mutants were also inactive in several reporter gene transactivation assays and in inhibition of PML-mediated repression. Importantly, a viral DNA genome containing an IE1 gene with a deletion [IE1(Delta290-320)] that was defective in these activities was not infectious when transfected into permissive fibroblast cells, but the mutant IE1(K450R), which is defective in IE1 sumoylation, remained infectious. Our mutational analysis strengthens the idea that interference by IE1 with both the sumoylation of PML and its repressor activity requires a physical interaction with PML that also leads to disruption of PODs. These activities of IE1 also correlate with several unusual transcriptional transactivation functions of IE1 and may be requirements for efficient initiation of the lytic cycle in vivo.


Asunto(s)
Citomegalovirus/patogenicidad , Fibroblastos/virología , Proteínas Inmediatas-Precoces/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Proteína SUMO-1/metabolismo , Factores de Transcripción/metabolismo , Proteínas Virales/metabolismo , Animales , Línea Celular , Células Cultivadas , Citomegalovirus/fisiología , Regulación de la Expresión Génica , Humanos , Proteínas Inmediatas-Precoces/genética , Proteína de la Leucemia Promielocítica , Transcripción Genética , Proteínas Supresoras de Tumor , Ubiquitina-Proteína Ligasas , Proteínas Virales/genética
20.
J Virol ; 78(9): 4847-65, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15078966

RESUMEN

The Epstein-Barr virus (EBV)-encoded ZTA protein interacts strongly with and stabilizes the cellular CCAAT/enhancer binding protein alpha (C/EBPalpha), leading to the induction of p21-mediated G(1) cell cycle arrest. Despite the strong interaction between these two basic leucine zipper (bZIP) family proteins, the ZTA and C/EBPalpha subunits do not heterodimerize, as indicated by an in vitro cross-linking assay with in vitro-cotranslated (35)S-labeled C/EBPalpha and (35)S-labeled ZTA protein. Instead, they evidently form a higher-order oligomeric complex that competes with C/EBPalpha binding but not with ZTA binding in electrophoretic mobility shift assays (EMSAs). Glutathione S-transferase affinity assays with mutant ZTA proteins revealed that the basic DNA binding domain and the key leucine zipper residues required for homodimerization are all required for the interaction with C/EBPalpha. ZTA is known to bind to two ZRE sites within the ZTA promoter and to positively autoregulate its own expression in transient cotransfection assays, but there is conflicting evidence about whether it does so in vivo. Examination of the proximal ZTA upstream promoter region by in vitro EMSA analysis revealed two high-affinity C/EBP binding sites (C-2 and C-3), which overlap the ZII and ZIIIB motifs, implicated as playing a key role in lytic cycle induction. A chromatin immunoprecipitation assay confirmed the in vivo binding of both endogenous C/EBPalpha and ZTA protein to the ZTA promoter after lytic cycle induction but not during the latent state in EBV-infected Akata cells. Reporter assays revealed that cotransfected C/EBPalpha activated the ZTA promoter even more effectively than cotransfected ZTA. However, synergistic activation of the ZTA promoter was not observed when ZTA and C/EBPalpha were cotransfected together in either HeLa or DG75 cells. Mutagenesis of either the ZII or the ZIIIB sites in the ZTA promoter strongly reduced C/EBPalpha transactivation, suggesting that these sites act cooperatively. Furthermore, the introduction of exogenous C/EBPalpha into EBV-infected HeLa-BX1 cells induced endogenous ZTA mRNA and protein expression, as demonstrated by both reverse transcription-PCR and immunoblotting assays. Finally, double-label immunofluorescence assays suggested that EAD protein expression was activated even better than ZTA expression in latently infected C/EBPalpha-transfected Akata cells, perhaps because of the presence of a strong B-cell-specific repressed chromatin conformation on the ZTA promoter itself during EBV latency.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Proteínas de Unión al ADN/metabolismo , Herpesvirus Humano 4/fisiología , Regiones Promotoras Genéticas/fisiología , Transactivadores/metabolismo , Activación Transcripcional , Proteínas Virales/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión , Chlorocebus aethiops , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Dimerización , Regulación Viral de la Expresión Génica , Células HeLa , Herpesvirus Humano 4/metabolismo , Humanos , Leucina Zippers , Datos de Secuencia Molecular , Unión Proteica , Subunidades de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Transactivadores/química , Transactivadores/genética , Transfección , Células Vero , Proteínas Virales/química , Proteínas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA