Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Virol ; 97(10): e0124123, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37772824

RESUMEN

IMPORTANCE: CD34+ hematopoietic progenitor cells (HPCs) are an important cellular reservoir for latent human cytomegalovirus (HCMV). Several HCMV genes are expressed during latency that are involved with the maintenance of the viral genome in CD34+ HPC. However, little is known about the process of viral reactivation in these cells. Here, we describe a viral protein, pUL8, and its interaction and stabilization with members of the Wnt/ß-catenin pathway as an important component of viral reactivation. We further define that pUL8 and ß-catenin interact with DVL2 via a PDZ-binding domain, and loss of UL8 interaction with ß-catenin-DVL2 restricts viral reactivation. Our findings will be instrumental in understanding the molecular processes involved in HCMV reactivation in order to design new antiviral therapeutics.


Asunto(s)
Antígenos CD34 , Citomegalovirus , Proteínas Dishevelled , Células Madre Hematopoyéticas , Proteínas Virales , Activación Viral , beta Catenina , Humanos , Antígenos CD34/metabolismo , beta Catenina/química , beta Catenina/metabolismo , Citomegalovirus/genética , Citomegalovirus/fisiología , Proteínas Dishevelled/química , Proteínas Dishevelled/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/virología , Dominios PDZ , Proteínas Virales/química , Proteínas Virales/metabolismo , Latencia del Virus/genética
2.
PLoS Pathog ; 17(8): e1009380, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34411201

RESUMEN

Human cytomegalovirus (HCMV) microRNAs (miRNAs) significantly rewire host signaling pathways to support the viral lifecycle and regulate host cell responses. Here we show that SMAD3 expression is regulated by HCMV miR-UL22A and contributes to the IRF7-mediated induction of type I IFNs and IFN-stimulated genes (ISGs) in human fibroblasts. Addition of exogenous TGFß interferes with the replication of a miR-UL22A mutant virus in a SMAD3-dependent manner in wild type fibroblasts, but not in cells lacking IRF7, indicating that downregulation of SMAD3 expression to limit IFN induction is important for efficient lytic replication. These findings uncover a novel interplay between SMAD3 and innate immunity during HCMV infection and highlight the role of viral miRNAs in modulating these responses.


Asunto(s)
Infecciones por Citomegalovirus/microbiología , Citomegalovirus/fisiología , Fibroblastos/microbiología , Inmunidad Innata/inmunología , Interferón Tipo I/metabolismo , MicroARNs/genética , Factor de Crecimiento Transformador beta/metabolismo , Fibroblastos/inmunología , Fibroblastos/patología , Interacciones Huésped-Patógeno , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Interferón Tipo I/genética , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/genética , Fenómenos Fisiológicos de los Virus
3.
Cell Host Microbe ; 27(1): 104-114.e4, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31866424

RESUMEN

Infection with human cytomegalovirus (HCMV) remains a significant cause of morbidity and mortality following hematopoietic stem cell transplant (HSCT) because of various hematologic problems, including myelosuppression. Here, we demonstrate that latently expressed HCMV miR-US5-2 downregulates the transcriptional repressor NGFI-A binding protein (NAB1) to induce myelosuppression of uninfected CD34+ hematopoietic progenitor cells (HPCs) through an increase in TGF-ß production. Infection of HPCs with an HCMVΔmiR-US5-2 mutant resulted in decreased TGF-ß expression and restoration of myelopoiesis. In contrast, we show that infected HPCs are refractory to TGF-ß signaling as another HCMV miRNA, miR-UL22A, downregulates SMAD3, which is required for maintenance of latency. Our data suggest that latently expressed viral miRNAs manipulate stem cell homeostasis by inducing secretion of TGF-ß while protecting infected HPCs from TGF-ß-mediated effects on viral latency and reactivation. These observations provide a mechanism through which HCMV induces global myelosuppression following HSCT while maintaining lifelong infection in myeloid lineage cells.


Asunto(s)
Citomegalovirus , Células Madre Hematopoyéticas/virología , MicroARNs/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Latencia del Virus , Antígenos CD34/metabolismo , Células Cultivadas , Citomegalovirus/genética , Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/metabolismo , Regulación hacia Abajo , Células HEK293 , Células Madre Hematopoyéticas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Células Mieloides/metabolismo , Células Mieloides/virología , Proteínas Represoras/metabolismo , Transducción de Señal , Proteína smad3/metabolismo , Activación Viral , Latencia del Virus/genética , Latencia del Virus/fisiología
4.
mBio ; 10(4)2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31431555

RESUMEN

Human cytomegalovirus (HCMV) infection of CD34+ hematopoietic progenitor cells (CD34+ HPCs) provides a critical reservoir of virus in stem cell transplant patients, and viral reactivation remains a significant cause of morbidity and mortality. The HCMV chemokine receptor US28 is implicated in the regulation of viral latency and reactivation. To explore the role of US28 signaling in latency and reactivation, we analyzed protein tyrosine kinase signaling in CD34+ HPCs expressing US28. US28-ligand signaling in CD34+ HPCs induced changes in key regulators of cellular activation and differentiation. In vitro latency and reactivation assays utilizing CD34+ HPCs indicated that US28 was required for viral reactivation but not latency establishment or maintenance. Similarly, humanized NSG mice (huNSG) infected with TB40E-GFP-US28stop failed to reactivate upon treatment with granulocyte-colony-stimulating factor, but viral genome levels were maintained. Interestingly, HCMV-mediated changes in hematopoiesis during latency in vivo and in vitro was also dependent upon US28, as US28 directly promoted differentiation toward the myeloid lineage. To determine whether US28 constitutive activity and/or ligand-binding activity were required for latency and reactivation, we infected both huNSG mice and CD34+ HPCs in vitro with HCMV TB40E-GFP containing the US28-R129A mutation (no CA) or Y16F mutation (no ligand binding). TB40E-GFP-US28-R129A was maintained during latency and exhibited normal reactivation kinetics. In contrast, TB40E-GFP-US28-Y16F exhibited high levels of viral genome during latency and reactivation, indicating that the virus did not establish latency. These data indicate that US28 is necessary for viral reactivation and ligand binding activity is required for viral latency, highlighting the complex role of US28 during HCMV latency and reactivation.IMPORTANCE Human cytomegalovirus (HCMV) can establish latency following infection of CD34+ hematopoietic progenitor cells (HPCs), and reactivation from latency is a significant cause of viral disease and accelerated graft failure in bone marrow and solid-organ transplant patients. The precise molecular mechanisms of HCMV infection in HPCs are not well defined; however, select viral gene products are known to regulate aspects of latency and reactivation. The HCMV-encoded chemokine receptor US28, which binds multiple CC chemokines as well as CX3CR1, is expressed both during latent and lytic phases of the virus life cycle and plays a role in latency and reactivation. However, the specific timing of US28 expression and the role of ligand binding in these processes are not well defined. In this report, we determined that US28 is required for reactivation but not for maintaining latency. However, when present during latency, US28 ligand binding activity is critical to maintaining the virus in a quiescent state. We attribute the regulation of both latency and reactivation to the role of US28 in promoting myeloid lineage cell differentiation. These data highlight the dynamic and multifunctional nature of US28 during HCMV latency and reactivation.


Asunto(s)
Antígenos CD34/metabolismo , Citomegalovirus/fisiología , Células Madre Hematopoyéticas/virología , Ligandos , Receptores de Quimiocina/metabolismo , Proteínas Virales/metabolismo , Latencia del Virus/fisiología , Animales , Diferenciación Celular , Citomegalovirus/genética , Citomegalovirus/patogenicidad , Genoma Viral , Hematopoyesis , Interacciones Huésped-Patógeno , Humanos , Ratones , Receptores de Quimiocina/genética , Transducción de Señal , Proteínas Virales/genética , Activación Viral/genética , Activación Viral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA