Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Virol ; 95(11)2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33731453

RESUMEN

HCMV establishes latency in myeloid cells. Using the Kasumi-3 latency model, we previously showed that lytic gene expression is activated prior to establishment of latency in these cells. The early events in infection may have a critical role in shaping establishment of latency. Here, we have used an integrative multi-omics approach to investigate dynamic changes in host and HCMV gene expression and epigenomes at early times post infection. Our results show dynamic changes in viral gene expression and viral chromatin. Analyses of Pol II, H3K27Ac and H3K27me3 occupancy of the viral genome showed that 1) Pol II occupancy was highest at the MIEP at 4 hours post infection. However, it was observed throughout the genome; 2) At 24 hours, H3K27Ac was localized to the major immediate early promoter/enhancer and to a possible second enhancer in the origin of replication OriLyt; 3) viral chromatin was broadly accessible at 24 hpi. In addition, although HCMV infection activated expression of some host genes, we observed an overall loss of de novo transcription. This was associated with loss of promoter-proximal Pol II and H3K27Ac, but not with changes in chromatin accessibility or a switch in modification of H3K27.Importance.HCMV is an important human pathogen in immunocompromised hosts and developing fetuses. Current anti-viral therapies are limited by toxicity and emergence of resistant strains. Our studies highlight emerging concepts that challenge current paradigms of regulation of HCMV gene expression in myeloid cells. In addition, our studies show that HCMV has a profound effect on de novo transcription and the cellular epigenome. These results may have implications for mechanisms of viral pathogenesis.

2.
Am J Transplant ; 19(9): 2421-2433, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30947382

RESUMEN

Reactivation of latent cytomegalovirus remains an important complication after transplant. Although immunosuppression (IS) has been implicated as a primary cause, we have previously shown that the implantation response of a kidney allograft can lead to early transcriptional activation of latent murine cytomegalovirus (MCMV) genes in an immune-competent host and to MCMV reactivation and dissemination to other organs in a genetically immune-deficient recipient. We now describe a model that allows us to separately analyze the impact of the implantation effect vs that of a clinically relevant IS regimen. Treatment with IS of latently infected mice alone does not induce viral reactivation, but transplant of latently infected allogeneic kidneys combined with IS facilitates MCMV reactivation in the graft and dissemination to other organs. The IS regimen effectively dampens allo-immune inflammatory pathways and depletes recipient anti-MCMV but does not affect ischemia-reperfusion injury pathways. MCMV reactivation similar to that seen in allogeneic transplants combined with also occurs after syngeneic transplants. Thus, our data strongly suggest that while ischemia-reperfusion injury of the implanted graft is sufficient and necessary to initiate transcriptional reactivation of latent MCMV ("first hit"), IS is permissive to the first hit and facilitates dissemination to other organs ("second hit").


Asunto(s)
Infecciones por Citomegalovirus/complicaciones , Trasplante de Riñón/efectos adversos , Muromegalovirus/fisiología , Insuficiencia Renal/cirugía , Activación Viral , Animales , Modelos Animales de Enfermedad , Eliminación de Gen , Histonas/metabolismo , Terapia de Inmunosupresión , Riñón/patología , Ratones , Ratones Endogámicos BALB C , Fenotipo , Complicaciones Posoperatorias/virología , Proteómica , Insuficiencia Renal/complicaciones , Daño por Reperfusión , Trasplante Homólogo
3.
J Gen Virol ; 97(4): 941-954, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26795571

RESUMEN

Reactivation of latent human cytomegalovirus is a significant infectious complication of organ transplantation and current therapies target viral replication once reactivation of latent virus has already occurred. The specific molecular pathways that activate viral gene expression in response to transplantation are not well understood. Our studies aim to identify these factors, with the goal of developing novel therapies that prevent transcriptional reactivation in transplant recipients. Murine cytomegalovirus (MCMV) is a valuable model for studying latency and reactivation of CMV in vivo. We previously demonstrated that transplantation of MCMV-latently infected kidneys into allogeneic recipients induces reactivation of immediate early (IE) gene expression and epigenetic reprogramming of the major immediate early promoter (MIEP) within 48 h. We hypothesize that these events are mediated by activation of signalling pathways that lead to binding of transcription factors to the MIEP, including AP-1 and NF-κB. Here we show that transplantation induces rapid activation of several members of the AP-1 and NF-κB transcription factor family and we demonstrate that canonical NF-κB (p65/p50), the junD component of AP-1, and nucleosome remodelling complexes are recruited to the MIEP following transplantation. Proteomic analysis of recipient plasma and transcriptome analysis of kidney RNA identified five extracellular ligands, including TNF, IL-1ß, IL-18, CD40L and IL-6, and three intracellular signalling pathways associated with reactivation of IE gene expression. Identification of the factors that mediate activation of these signalling pathways may eventually lead to new therapies to prevent reactivation of CMV and its sequelae.


Asunto(s)
Infecciones por Herpesviridae/genética , Proteínas Inmediatas-Precoces/genética , Trasplante de Riñón , Muromegalovirus/genética , FN-kappa B/genética , Factor de Transcripción AP-1/genética , Activación Viral , Animales , Ligando de CD40/genética , Ligando de CD40/inmunología , Femenino , Regulación de la Expresión Génica/inmunología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/patología , Interacciones Huésped-Patógeno , Proteínas Inmediatas-Precoces/inmunología , Interleucina-18/genética , Interleucina-18/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Muromegalovirus/inmunología , FN-kappa B/inmunología , Nucleosomas/genética , Nucleosomas/inmunología , Regiones Promotoras Genéticas , Proteoma/genética , Proteoma/inmunología , Transducción de Señal , Factor de Transcripción AP-1/inmunología , Trasplante Homólogo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/inmunología , Latencia del Virus
4.
Microbiol Spectr ; 11(1): e0314422, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-36645269

RESUMEN

Human cytomegalovirus (HCMV) is an opportunistic pathogen that infects most of the population. The complex 236 kbp genome encodes more than 170 open reading frames, whose expression is temporally regulated by both viral transcriptional regulators and cellular factors that control chromatin and transcription. Here, we have used state of the art genomic technologies to investigate the viral transcriptome in conjunction with 2 key transcriptional regulators: Pol II and H3K27Ac. Although it is well known that the major immediate early (IE) proteins activate early gene expression through both direct and indirect interactions, and that histone modifications play an important role in regulating viral gene expression, the role of the IE proteins in modulating viral chromatin is not fully understood. To address this question, we have used a virus engineered for conditional expression of the IE proteins combined with RNA and Chromatin immunoprecipitation (ChIP) analyses to assess the role of these proteins in modulating both viral chromatin and gene expression. Our results show that (i) there is an enhancer-like element in OriLyt that is extraordinarily enriched in H3K27Ac; (ii) in addition to activation of viral gene expression, the IE proteins play a critical role in recruitment of Pol II and H3K27Ac to this element. IMPORTANCE HCMV is an important human pathogen associated with complications in transplant patients and birth defects. The complex program of viral gene expression is regulated by both viral proteins and host factors. Here, we have investigated the role of the immediate early proteins in regulating the viral epigenome. Our results show that the viral immediate early proteins bring about an enormous enrichment of H3K27Ac marks at the OriLyt RNA4.9 promoter, concomitant with an increase in RNA4.9 expression. This epigenetic characteristic adds importantly to the view that OriLyt has structural and functional characteristics of a strong enhancer that, we now discover, is regulated by IE proteins.


Asunto(s)
Proteínas Inmediatas-Precoces , Humanos , Proteínas Inmediatas-Precoces/genética , Citomegalovirus/genética , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Cromatina/genética , Regulación Viral de la Expresión Génica
5.
J Virol ; 84(7): 3631-43, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20106920

RESUMEN

Our previous studies showed that establishment of murine cytomegalovirus (MCMV) latency in vivo is associated with repression of immediate-early gene expression, deacetylation of histones bound to the major immediate-early promoter (MIEP), changes in patterns of methylation of histones, and recruitment of cellular repressors of transcription to the MIEP. Here, we have quantitatively analyzed the kinetics of changes in viral RNA expression, DNA copy number, and recruitment of repressors and activators of transcription to viral promoters during the course of infection. Our results show that changes in viral gene expression correlate with changes in recruitment of RNA polymerase and acetylated histones to viral promoters. Binding of the transcriptional repressors histone deacetylase type 2 (HDAC2), HDAC3, YY1, CBF-1/RBP-Jk, Daxx, and CIR to the MIEP and HDACs to other promoters showed a biphasic pattern: some binding was detectable prior to activation of viral gene expression, then decreased with the onset of transcription and increased again as repression of viral gene expression occurred. Potential binding sites for CBF-1/RBP-Jk and YY1 in the MIEP and for YY1 in the M100 promoter (M100P) were identified by in silico analysis. While recruitment of HDACs was not promoter specific, binding of CBF-1/RBP-Jk and YY1 was restricted to promoters with their cognate sites. Our results suggest that sequences within viral promoters may contribute to establishment of latency through recruitment of transcriptional repressors to these genes. The observation that repressors are bound to the MIEP and other promoters immediately upon infection suggests that latency may be established in some cells very early in infection.


Asunto(s)
Genes Inmediatos-Precoces , Infecciones por Herpesviridae/metabolismo , Muromegalovirus/genética , Regiones Promotoras Genéticas , Proteínas Represoras/metabolismo , Acetilación , Animales , Femenino , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Ratones , Ratones Endogámicos BALB C , Activación Transcripcional , Factor de Transcripción YY1/metabolismo
6.
Artículo en Inglés | MEDLINE | ID: mdl-32296651

RESUMEN

CMV is an ancient herpesvirus that has co-evolved with its host over millions of years. The 236 kbp genome encodes at least 165 genes, four non-coding RNAs and 14 miRNAs. Of the protein-coding genes, 43-44 are core replication genes common to all herpesviruses, while ~30 are unique to betaherpesviruses. Many CMV genes are involved in evading detection by the host immune response, and others have roles in cell tropism. CMV replicates systemically, and thus, has adapted to various biological niches within the host. Different biological niches may place competing demands on the virus, such that genes that are favorable in some contexts are unfavorable in others. The outcome of infection is dependent on the cell type. In fibroblasts, the virus replicates lytically to produce infectious virus. In other cell types, such as myeloid progenitor cells, there is an initial burst of lytic gene expression, which is subsequently silenced through epigenetic repression, leading to establishment of latency. Latently infected monocytes disseminate the virus to various organs. Latency is established through cell type specific mechanisms of transcriptional silencing. In contrast, reactivation is triggered through pathways activated by inflammation, infection, and injury that are common to many cell types, as well as differentiation of myeloid cells to dendritic cells. Thus, CMV has evolved a complex relationship with the host immune response, in which it exploits cell type specific mechanisms of gene regulation to establish latency and to disseminate infection systemically, and also uses the inflammatory response to infection as an early warning system which allows the virus to escape from situations in which its survival is threatened, either by cellular damage or infection of the host with another pathogen. Spontaneous reactivation induced by cellular aging/damage may explain why extensive expression of lytic genes has been observed in recent studies using highly sensitive transcriptome analyses of cells from latently infected individuals. Recent studies with animal models highlight the potential for harnessing the host immune response to blunt cellular injury induced by organ transplantation, and thus, prevent reactivation of CMV and its sequelae.


Asunto(s)
Citomegalovirus , Latencia del Virus , Animales , Citomegalovirus/genética , Expresión Génica , Humanos , Inmunidad , Células Mieloides , Activación Viral
7.
Transplantation ; 104(5): e118-e124, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31996662

RESUMEN

Cytomegalovirus (CMV) is a ß-herpesvirus that establishes lifelong latency in infected hosts. Following transplantation of a latently infected organ, reactivation can occur and consists of a spectrum of clinically apparent syndromes from mild symptoms to tissue-invasive, resulting in both direct and indirect sequelae. Before the advent of effective antiviral agents, the primary treatment was reduction in immunosuppression (IS). While antiviral agents provide effective prophylaxis, there are several important caveats associated with their use, including drug toxicity and resistance. The traditional view attributes CMV reactivation and the ensuing clinical disease primarily to IS, either intrinsic to disease-related immune compromise or from the extrinsic administration of IS agents. However, previous data from both animal models and human subjects showed that inflammatory signals could induce upregulation of latent viral gene expression. New data demonstrate that ischemia/reperfusion is necessary and sufficient to induce CMV reactivation following murine transplantation of a latently infected graft. In this article, we review a growing body of evidence that suggests that reactivation of both human CMV and murine CMV is first triggered by molecular events that activate CMV gene expression and lytic infection and viral dissemination are then facilitated by IS. The initial activation of viral gene expression may be mediated by oxidative stress, DNA damage, or inflammatory cytokines, and these factors may act synergistically. New therapeutic approaches are needed to capture this complex array of targets.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Rechazo de Injerto/inmunología , Inmunosupresores/uso terapéutico , Trasplante de Riñón , Activación Viral/inmunología , Latencia del Virus/inmunología , Animales , Anticuerpos Antivirales/inmunología , Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/virología , Rechazo de Injerto/prevención & control , Humanos
8.
Pathogens ; 9(8)2020 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-32722544

RESUMEN

Transplantation tolerance is achieved when recipients are unresponsive to donor alloantigen yet mobilize against third-party antigens, including virus. After transplantation, cytomegalovirus (CMV) reactivation in latently-infected transplants reduces allograft viability. To determine if pre-tolerized recipients are resistant to viral dissemination in this setting, we transfused chemically-fixed donor splenocytes (1-ethyl-3- (3'-dimethyl-aminopropyl)-carbo-diimide (ECDI)-treated splenocytes (ECDIsp)) to induce donor antigen tolerance without immunosuppression. In parallel, we implanted donor islet cells to validate operational tolerance. These pre-tolerized recipients were implanted with murine CMV (MCMV) latently-infected donor kidneys (a validated model of CMV latency) to monitor graft inflammation and viral dissemination. Our results indicate that tolerance to donor islets was sustained in recipients after implantation of donor kidneys. In addition, kidney allografts implanted after ECDIsp and islet implantation exhibited low levels of fibrosis and tubulitis. In contrast, kidney cellular and innate immune infiltrates trended higher in the CMV group and exhibited increased markers of CD8+ T cell activation. Tolerance induction was unable to prevent increases in MCMV-specific CD8+ T cells or dissemination of viral IE-1 DNA. Our data suggest that latently-infected allografts are inherently more susceptible to inflammation that is associated with viral dissemination in pre-tolerized recipients. Thus, CMV latently-infected allografts require enhanced strategies to protect allograft integrity and viral spread.

9.
J Virol ; 82(21): 10922-31, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18753203

RESUMEN

Human cytomegalovirus (CMV) is a ubiquitous herpesvirus with the ability to establish a lifelong latent infection. The mechanism by which this occurs is not well understood. Regulation of, for example, immediate-early (IE) gene expression is thought to be a critical control point in transcriptional control of the switch between latency and reactivation. Here, we present evidence that supports previous studies showing that the majority of genomes are quiescent with respect to gene expression. To study the possible role of epigenetic factors that may be involved in repression of ie gene expression in latency, we have analyzed changes in the patterns of modifications of histones bound to the major IE promoter (MIEP) in the kidneys of acutely and latently infected mice. Our studies show that, like herpes simplex virus, murine CMV genomes become relatively enriched in histones in latent infection. There are dramatic changes in modifications of histones associated with the MIEP when latency is established: H3 and H4 become hypoacetylated and H3 is hypomethylated at lysine 4, while H3 lysine 9 is hypermethylated in latently infected mice. These changes are accompanied by a relative loss of RNA polymerase and gain of heterochromatin protein 1gamma and Yin-Yang 1 bound to the MIEP. Our studies suggest that, in the majority of cells, CMV establishes a true latent infection, defined as the lack of expression of genes associated with productive infection, and that this occurs through changes in histone modifications and recruitment of transcriptional silencing factors to the MIEP.


Asunto(s)
ADN Viral/metabolismo , Genes Inmediatos-Precoces , Histonas/metabolismo , Muromegalovirus/fisiología , Regiones Promotoras Genéticas , Proteínas Represoras/metabolismo , Latencia del Virus , Acetilación , Animales , Femenino , Riñón/virología , Metilación , Ratones , Ratones Endogámicos BALB C
10.
Transplantation ; 85(7): 1039-45, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18408586

RESUMEN

BACKGROUND: Reactivation of latent human cytomegalovirus (HCMV) infection is a significant risk factor for long term allograft dysfunction. The molecular pathways involved in reactivation of latent virus have not been identified. Previous studies suggested that tumor necrosis factor (TNF) -mediated activation of nuclear factor of kappa light polypeptide gene enhancer in B-cells (NF-kappa B) leading to transcriptional reactivation of viral immediate early (ie) gene expression might be important in transplant-associated viral reactivation. CMV IE gene expression is controlled by the major immediate early promoter/enhancer (MIEP). Because HCMV does not infect mice, transgenic mice carrying a beta-galactosidase reporter gene under the control of the HCMV immediate early enhancer (MIEP-lacZ mice) are a valuable model for studying regulation of CMV IE gene expression in vivo. We have used TNF receptor-deficient MIEP-lacZ (MIEP-lacZ TNFR DKO) mice to study the requirement for TNF in transplant-induced activation of the MIEP. METHODS: Allogenic kidney transplants were performed using MIEP-lacZ TNFR DKO or MIEP-lacZ TNFRwild-type donor mice. beta-Galactosidase activity was used to measure activation of the IE enhancer in donor kidneys at 2 days of posttransplantation and in contralateral controls. Transcription factor activation was assayed with Trans-Am kits. RESULTS: Allogenic and syngenic transplantation activate the HCMV IE enhancer to the same extent. TNF receptor signaling was not required for activation of the MIEP. TNF receptor signaling was required for activation of NF-kappaB, but not for activation of activating protein 1 family members junD and Fra-1 in day 2 allografts. CONCLUSIONS: TNF-independent pathways can activate the enhancer in response to allogenic transplantation. This may occur through activation of MIEP-binding transcription factors other than NF-kappa B.


Asunto(s)
Citomegalovirus/genética , Trasplante de Riñón/fisiología , Receptores Tipo I de Factores de Necrosis Tumoral/fisiología , Receptores del Factor de Necrosis Tumoral/fisiología , Activación Viral/fisiología , Animales , Citomegalovirus/fisiología , Elementos de Facilitación Genéticos , Riñón/enzimología , Ratones , Ratones Noqueados , FN-kappa B/fisiología , Receptores del Factor de Necrosis Tumoral/deficiencia , Receptores del Factor de Necrosis Tumoral/genética , Transducción de Señal , Trasplante Homólogo , beta-Galactosidasa/metabolismo
11.
mBio ; 9(5)2018 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-30206173

RESUMEN

We used the Kasumi-3 model to study human cytomegalovirus (HCMV) latency and reactivation in myeloid progenitor cells. Kasumi-3 cells were infected with HCMV strain TB40/Ewt-GFP, flow sorted for green fluorescent protein-positive (GFP+) cells, and cultured for various times to monitor establishment of latency, as judged by repression of viral gene expression (RNA/DNA ratio) and loss of virus production. We found that, in the vast majority of cells, latency was established posttranscriptionally in the GFP+ infected cells: transcription was initially turned on and then turned off. We also found that some of the GFP- cells were infected, suggesting that latency might be established in these cells at the outset of infection. We were not able to test this hypothesis because some GFP- cells expressed lytic genes and thus it was not possible to separate them from GFP- quiescent cells. In addition, we found that the pattern of expression of lytic genes that have been associated with latency, including UL138, US28, and RNA2.7, was the same as that of other lytic genes, indicating that there was no preferential expression of these genes once latency was established. We confirmed previous studies showing that tumor necrosis factor alpha (TNF-α) induced reactivation of infectious virus, and by analyzing expression of the progenitor cell marker CD34 as well as myeloid cell differentiation markers in IE+ cells after treatment with TNF-α, we showed that TNF-α induced transcriptional reactivation of IE gene expression independently of differentiation. TNF-α-mediated reactivation in Kasumi-3 cells was correlated with activation of NF-κB, KAP-1, and ATM.IMPORTANCE HCMV is an important human pathogen that establishes lifelong latent infection in myeloid progenitor cells and reactivates frequently to cause significant disease in immunocompromised people. Our observation that viral gene expression is first turned on and then turned off to establish latency suggests that there is a host defense, which may be myeloid cell specific, responsible for transcriptional silencing of viral gene expression. Our observation that TNF-α induces reactivation independently of differentiation provides insight into molecular mechanisms that control reactivation.


Asunto(s)
Diferenciación Celular , Citomegalovirus/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Activación Viral/efectos de los fármacos , Latencia del Virus , Antígenos CD34/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular Tumoral , Citomegalovirus/genética , Citomegalovirus/fisiología , Citometría de Flujo , Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células Mieloides/virología , FN-kappa B/metabolismo , Procesamiento Postranscripcional del ARN , Proteína 28 que Contiene Motivos Tripartito/metabolismo
12.
Artículo en Inglés | MEDLINE | ID: mdl-28707002

RESUMEN

Myeloid cells are important cell types that carry human cytomegalovirus. Latent viral DNA is present in CD34+ progenitor cells and their derived monocytes. However, differentiation of latently infected monocytes to mature macrophages or dendritic cells causes reactivation of latent viruses. During hematopoietic development, pluripotent genes are repressed, and lineage specific genes are activated in a step-wise manner. This process is governed by cell-type specific chromatin states. Enhancers in the hematopoietic system are highly dynamic and established by pioneer (first tier) transcription factors (TFs), which set the stage for second and third tier TF binding. In this review, we examine the epigenetic mechanisms that regulate myeloid cell development, cell identity, and activation with a special focus on factors that regulate viral gene expression and the status of viral infection in myeloid cells.

13.
J Clin Virol ; 25 Suppl 2: S123-36, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12361763

RESUMEN

BACKGROUND: Reactivation of CMV from latency results in serious morbidity and mortality in immunocompromised transplant recipients. The mechanism by which CMV reactivates from latency has not been well understood. OBJECTIVE: In this review we discuss three models for reactivation from latency and present evidence in favor of the model that reactivation is a multi-step process which is initiated by the allogeneic response to the transplanted organ. Study design (J. Virol. 75 (2001) 4814). Mice latently infected with murine cytomegalovirus (MCMV) were used as donors for allogeneic or syngeneic kidney transplants into immunocompetent recipients. The contralateral donor kidneys were used as controls. Transplanted kidneys were removed at various times after transplant and analyzed for expression of viral genes associated with productive infection and for expression of inflammatory cytokines. Electrophoretic mobility shift assay was performed on nuclear extracts of control and transplanted kidneys to examine activation of AP-1 and NFkappaB. Latently infected mice were also injected with tumor necrosis factor (TNF) to examine the effect of TNF alone on induction of MCMV immediate-early (IE) gene expression. Transgenic major immediate early promoter-lacZ mice carrying a beta-galactosidase reporter gene under the control of the human cytomegalovirus (HCMV) IE promoter/enhancer were used as donors for allogeneic kidney transplants to study the effect of allogeneic transplantation on induction of HCMV IE gene expression. RESULTS: Allogeneic, but not syngeneic transplantation induces MCMV IE-1 expression and expression of inflammatory cytokines, including TNF. Allogeneic transplantation activates transcription factors, including NFkappaB and AP-1. TNF alone can induce MCMV IE-1 gene expression and activation of NFkappaB and AP-1 in some tissues. CONCLUSIONS: We propose that induction of IE-1 gene expression is the first step in reactivation of the virus in an immunocompromised transplant recipient, and that it occurs as a result of the allogeneic response, which induces expression of TNF and subsequent activation of NFkappaB, and ischemia/reperfusion injury, which induces activation of AP-1. We speculate that the natural stimulus for reactivation in an immunocompetent host is an inflammatory immune response to infection and that allogeneic transplantation mimics this process.


Asunto(s)
Citomegalovirus/fisiología , Modelos Animales de Enfermedad , Trasplante de Riñón/efectos adversos , Trasplante Homólogo/efectos adversos , Activación Viral , Latencia del Virus , Animales , Línea Celular , Infecciones por Citomegalovirus/virología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , FN-kappa B/metabolismo
14.
Viruses ; 5(5): 1325-45, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23698401

RESUMEN

Cytomegalovirus (CMV) gene expression is repressed in latency due to heterochromatinization of viral genomes. In murine CMV (MCMV) latently infected mice, viral genomes are bound to histones with heterochromatic modifications, to enzymes that mediate these modifications, and to adaptor proteins that may recruit co-repressor complexes. Kinetic analyses of repressor binding show that these repressors are recruited at the earliest time of infection, suggesting that latency may be the default state. Kidney transplantation leads to epigenetic reprogramming of latent viral chromatin and reactivation of immediate early gene expression. Inflammatory signaling pathways, which activate transcription factors that regulate the major immediate early promoter (MIEP), likely mediate the switch in viral chromatin.


Asunto(s)
Citomegalovirus/fisiología , Epigénesis Genética , Regulación Viral de la Expresión Génica , Interacciones Huésped-Patógeno , Activación Viral , Latencia del Virus , Citomegalovirus/genética , Humanos
15.
Transplantation ; 87(2): 178-88, 2009 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-19155971

RESUMEN

BACKGROUND: Increased levels of tumor necrosis factor (TNF) are a risk factor for allograft rejection. In vitro studies have shown that binding of TNF to its receptor activates signaling cascades that induce expression of many genes involved in inflammation. The role of intragraft TNF receptor (TNFR) signaling in activation of gene expression in allografts has not been studied. METHODS: Gene expression profiling and quantitative real-time polymerase chain reaction analysis were used to investigate the role of TNFR signaling in the early intragraft activation of cellular gene expression in renal allografts at 2 days posttransplant. RESULTS: The TNFRs play a critical role in activating intragraft expression of transcription factors controlling innate and adaptive immunity and stress responses (interferon regulatory factor [IRF]1, IRF 8, Isgf3g, and ATF3) of cytokines and receptors mediating inflammation (TNF, interleukin [IL]-6, interferon-gamma, oncostatin M receptor [OMCR], toll-like receptor [TLR]2, and IL-2Rgamma), of chemokines and adhesion molecules that recruit inflammatory cells (Cxcl9, Cxcl11, E-selectin, and intracellular adhesion molecule [ICAM]-1), of genes involved in costimulation of T cells and processing and presentation of antigens (H2-DMb, Psmb8, and CD40), and genes that mediate the response to interferons. In addition to its proinflammatory role, TNFR signaling induces expression of SOCS3, a negative regulator of IL-6 and OSMR signaling and Nfkbie, and a negative regulator of TNFR signal transduction. CONCLUSIONS: These studies illustrate the pleiotropic effect of TNF in both activation and down-modulation of the immune response and the complex interactions between the TNFRs and other cytokine signaling pathways in the early allograft response.


Asunto(s)
Inmunidad Innata , Inflamación/inmunología , Trasplante de Riñón/inmunología , Riñón/inmunología , Riñón/cirugía , Receptores del Factor de Necrosis Tumoral/inmunología , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Citocinas/genética , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Inmunidad Innata/genética , Inflamación/genética , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Receptores del Factor de Necrosis Tumoral/genética , Reproducibilidad de los Resultados , Transducción de Señal/genética , Factores de Tiempo , Factores de Transcripción/genética , Trasplante Homólogo
16.
J Gen Virol ; 88(Pt 4): 1097-1102, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17374752

RESUMEN

We have used a spleen explant model to investigate mechanisms of murine cytomegalovirus latency and reactivation. Induction of immediate-early (ie) gene expression occurs in explants after approximately 9 days in culture and virus reactivation follows induction of ie gene expression with kinetics similar to that of productive infection in vitro. This occurs independently of TNF receptor signalling. Treatment with the DNA methylation inhibitor 5-aza-2'-deoxycytidine and the histone deacetylase inhibitor trichostatin A results in more rapid induction of ie gene expression and reactivation of virus. Despite these results, which suggest a role for DNA methylation in maintenance of viral latency, we find that the major immediate-early promoter/enhancer is not methylated in latently infected mice. Our results support the hypothesis that latency is maintained by epigenetic control of ie gene expression, and that induction of ie gene expression leads to reactivation of virus, but suggest that these are not controlled by DNA methylation.


Asunto(s)
Azacitidina/análogos & derivados , Regulación Viral de la Expresión Génica/efectos de los fármacos , Genes Inmediatos-Precoces/efectos de los fármacos , Ácidos Hidroxámicos/farmacología , Muromegalovirus/efectos de los fármacos , Provirus/efectos de los fármacos , Activación Viral/efectos de los fármacos , Animales , Azacitidina/farmacología , Metilación de ADN , ADN Viral/química , Decitabina , Inhibidores Enzimáticos/farmacología , Proteínas Inmediatas-Precoces/biosíntesis , Ratones , Modelos Animales , Muromegalovirus/genética , Muromegalovirus/fisiología , Regiones Promotoras Genéticas , Provirus/genética , Provirus/fisiología , Transcripción Genética
17.
Am J Transplant ; 5(7): 1606-13, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15943618

RESUMEN

Reactivation of latent human cytomegalovirus is of significant concern in immunocompromised transplant patients and is likely to occur through transcriptional activation of immediate early (ie) gene expression through mechanisms that are not well understood. TNF-mediated activation of NF-kappaB has been proposed to be one pathway leading to transcriptional activation of CMV ie gene expression. Using transgenic mice carrying a lacZ reporter gene under the control of the HCMV major ie promoter/enhancer (MIEP-lacZ mice) and MIEP-lacZ mice deficient in TNF receptor 1 and TNF receptor 2 (MIEP-lac Z TNFR DKO mice), we demonstrate that renal ischemia/reperfusion (I/R) injury activates the HCMV enhancer independently of TNF. Induction of MIEP-lacZ expression was preceded by TNFR-independent formation of reactive oxygen species (ROS), weak and transient activation of NF-kappaB and strong and sustained activation of AP-1. Our studies show that, in addition to TNF-mediated signaling, TNF-independent signaling induced by I/R injury can contribute to the activation of the HCMV enhancer. This likely occurs through ROS-mediated activation of AP-1. Targeting MAP kinase signaling pathways as well as NF-kappaB may be of therapeutic value in patients with CMV infection.


Asunto(s)
Antígenos Virales/genética , Citomegalovirus/genética , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Proteínas Inmediatas-Precoces/genética , Riñón/irrigación sanguínea , Regiones Promotoras Genéticas/genética , Daño por Reperfusión/virología , Animales , Proteína de Unión a CREB , Ratones , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Receptores Tipo II del Factor de Necrosis Tumoral/deficiencia , Daño por Reperfusión/metabolismo , Transactivadores/metabolismo , Factor de Transcripción AP-1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA