Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Semin Cell Dev Biol ; 146: 2-19, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36463091

RESUMEN

Viruses have evolved a multitude of mechanisms to combat barriers to productive infection in the host cell. Virally-encoded miRNAs are one such means to regulate host gene expression in ways that benefit the virus lifecycle. miRNAs are small non-coding RNAs that regulate protein expression but do not trigger the adaptive immune response, making them powerful tools encoded by viruses to regulate cellular processes. Diverse viruses encode for miRNAs but little sequence homology exists between miRNAs of different viral species. Despite this, common cellular pathways are targeted for regulation, including apoptosis, immune evasion, cell growth and differentiation. Herein we will highlight the viruses that encode miRNAs and provide mechanistic insight into how viral miRNAs aid in lytic and latent infection by targeting common cellular processes. We also highlight how viral miRNAs can mimic host cell miRNAs as well as how viral miRNAs have evolved to regulate host miRNA expression. These studies dispel the myth that viral miRNAs are subtle regulators of gene expression, and highlight the critical importance of viral miRNAs to the virus lifecycle.


Asunto(s)
MicroARNs , Virus , MicroARNs/genética , MicroARNs/metabolismo , Virus/genética , Virus/metabolismo , Diferenciación Celular , Procesamiento Proteico-Postraduccional , Expresión Génica , Regulación Viral de la Expresión Génica/genética , Regulación de la Expresión Génica
2.
PLoS Pathog ; 19(10): e1011682, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37782657

RESUMEN

Human cytomegalovirus (HCMV) encodes multiple putative G protein-coupled receptors (GPCRs). US28 functions as a viral chemokine receptor and is expressed during both latent and lytic phases of virus infection. US28 actively promotes cellular migration, transformation, and plays a major role in mediating viral latency and reactivation; however, knowledge about the interaction partners involved in these processes is still incomplete. Herein, we utilized a proximity-dependent biotinylating enzyme (TurboID) to characterize the US28 interactome when expressed in isolation, and during both latent (CD34+ hematopoietic progenitor cells) and lytic (fibroblasts) HCMV infection. Our analyses indicate that the US28 signalosome converges with RhoA and EGFR signal transduction pathways, sharing multiple mediators that are major actors in processes such as cellular proliferation and differentiation. Integral members of the US28 signaling complex were validated in functional assays by immunoblot and small-molecule inhibitors. Importantly, we identified RhoGEFs as key US28 signaling intermediaries. In vitro latency and reactivation assays utilizing primary CD34+ hematopoietic progenitor cells (HPCs) treated with the small-molecule inhibitors Rhosin or Y16 indicated that US28 -RhoGEF interactions are required for efficient viral reactivation. These findings were recapitulated in vivo using a humanized mouse model where inhibition of RhoGEFs resulted in a failure of the virus to reactivate. Together, our data identifies multiple new proteins in the US28 interactome that play major roles in viral latency and reactivation, highlights the utility of proximity-sensor labeling to characterize protein interactomes, and provides insight into targets for the development of novel anti-HCMV therapeutics.


Asunto(s)
Citomegalovirus , Transducción de Señal , Animales , Ratones , Humanos , Citomegalovirus/fisiología , Latencia del Virus , Diferenciación Celular , Células Madre Hematopoyéticas
3.
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
4.
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
5.
PLoS Pathog ; 17(1): e1009255, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33508041

RESUMEN

Cytomegalovirus (CMV) causes clinically important diseases in immune compromised and immune immature individuals. Based largely on work in the mouse model of murine (M)CMV, there is a consensus that myeloid cells are important for disseminating CMV from the site of infection. In theory, such dissemination should expose CMV to cell-mediated immunity and thus necessitate evasion of T cells and NK cells. However, this hypothesis remains untested. We constructed a recombinant MCMV encoding target sites for the hematopoietic specific miRNA miR-142-3p in the essential viral gene IE3. This virus disseminated poorly to the salivary gland following intranasal or footpad infections but not following intraperitoneal infection in C57BL/6 mice, demonstrating that dissemination by hematopoietic cells is essential for specific routes of infection. Remarkably, depletion of NK cells or T cells restored dissemination of this virus in C57BL/6 mice after intranasal infection, while dissemination occurred normally in BALB/c mice, which lack strong NK cell control of MCMV. These data show that cell-mediated immunity is responsible for restricting MCMV to hematopoietic cell-mediated dissemination. Infected hematopoietic cells avoided cell-mediated immunity via three immune evasion genes that modulate class I MHC and NKG2D ligands (m04, m06 and m152). MCMV lacking these 3 genes spread poorly to the salivary gland unless NK cells were depleted, but also failed to replicate persistently in either the nasal mucosa or salivary gland unless CD8+ T cells were depleted. Surprisingly, CD8+ T cells primed after intranasal infection required CD4+ T cell help to expand and become functional. Together, our data suggest that MCMV can use both hematopoietic cell-dependent and -independent means of dissemination after intranasal infection and that cell mediated immune responses restrict dissemination to infected hematopoietic cells, which are protected from NK cells during dissemination by viral immune evasion. In contrast, viral replication within mucosal tissues depends on evasion of T cells.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Evasión Inmune , Inmunidad Celular , Muromegalovirus/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/virología , Infecciones por Herpesviridae/virología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Muromegalovirus/genética , Muromegalovirus/fisiología , Replicación Viral
6.
J Virol ; 95(3)2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33177198

RESUMEN

In human cytomegalovirus (HCMV)-seropositive patients, CD34+ hematopoietic progenitor cells (HPCs) provide an important source of latent virus that reactivates following cellular differentiation into tissue macrophages. Multiple groups have used primary CD34+ HPCs to investigate mechanisms of viral latency. However, analyses of mechanisms of HCMV latency have been hampered by the genetic variability of CD34+ HPCs from different donors, availability of cells, and low frequency of reactivation. In addition, multiple progenitor cell types express surface CD34, and the frequencies of these populations differ depending on the tissue source of the cells and culture conditions in vitro In this study, we generated CD34+ progenitor cells from two different embryonic stem cell (ESC) lines, WA01 and WA09, to circumvent limitations associated with primary CD34+ HPCs. HCMV infection of CD34+ HPCs derived from either WA01 or WA09 ESCs supported HCMV latency and induced myelosuppression similar to infection of primary CD34+ HPCs. Analysis of HCMV-infected primary or ESC-derived CD34+ HPC subpopulations indicated that HCMV was able to establish latency and reactivate in CD38+ CD90+ and CD38+/low CD90- HPCs but persistently infected CD38- CD90+ cells to produce infectious virus. These results indicate that ESC-derived CD34+ HPCs can be used as a model for HCMV latency and that the virus either latently or persistently infects specific subpopulations of CD34+ cells.IMPORTANCE Human cytomegalovirus infection is associated with severe disease in transplant patients and understanding how latency and reactivation occur in stem cell populations is essential to understand disease. CD34+ hematopoietic progenitor cells (HPCs) are a critical viral reservoir; however, these cells are a heterogeneous pool with donor-to-donor variation in functional, genetic, and phenotypic characteristics. We generated a novel system using embryonic stem cell lines to model HCMV latency and reactivation in HPCs with a consistent cellular background. Our study defined three key stem cell subsets with differentially regulated latent and replicative states, which provide cellular candidates for isolation and treatment of transplant-mediated disease. This work provides a direction toward developing strategies to control the switch between latency and reactivation.


Asunto(s)
Antígenos CD34/metabolismo , Infecciones por Citomegalovirus/virología , Citomegalovirus/aislamiento & purificación , Células Madre Hematopoyéticas/virología , Interacciones Huésped-Patógeno , Células Madre Embrionarias Humanas/virología , Activación Viral , Latencia del Virus , Células Cultivadas , Infecciones por Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/patología , Células Madre Hematopoyéticas/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Humanos , Transducción de Señal
7.
PLoS Pathog ; 15(11): e1007854, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31725809

RESUMEN

Reactivation of latent Human Cytomegalovirus (HCMV) in CD34+ hematopoietic progenitor cells (HPCs) is closely linked to hematopoiesis. Viral latency requires maintenance of the progenitor cell quiescence, while reactivation initiates following mobilization of HPCs to the periphery and differentiation into CD14+ macrophages. Early growth response gene 1 (EGR-1) is a transcription factor activated by Epidermal growth factor receptor (EGFR) signaling that is essential for the maintenance of CD34+ HPC self-renewal in the bone marrow niche. Down-regulation of EGR-1 results in mobilization and differentiation of CD34+ HPC from the bone marrow to the periphery. In the current study we demonstrate that the transcription factor EGR-1 is directly targeted for down-regulation by HCMV miR-US22 that results in decreased proliferation of CD34+ HPCs and a decrease in total hematopoietic colony formation. We also show that an HCMV miR-US22 mutant fails to reactivate in CD34+ HPCs, indicating that expression of EGR-1 inhibits viral reactivation. Since EGR-1 promotes CD34+ HPC self-renewal in the bone marrow niche, HCMV miR-US22 down-regulation of EGR-1 is a necessary step to block HPC self-renewal and proliferation to induce a cellular differentiation pathway necessary to promote reactivation of virus.


Asunto(s)
Proliferación Celular , Infecciones por Citomegalovirus/virología , Citomegalovirus/fisiología , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Células Madre Hematopoyéticas/citología , MicroARNs/genética , Activación Viral , Diferenciación Celular , Células Cultivadas , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Hematopoyesis , Células Madre Hematopoyéticas/virología , Interacciones Huésped-Patógeno , Humanos , Transducción de Señal
8.
Curr Top Microbiol Immunol ; 419: 243-280, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28674945

RESUMEN

Non-coding RNAs (ncRNAs) play essential roles in multiple aspects of the life cycles of herpesviruses and contribute to lifelong persistence of herpesviruses within their respective hosts. In this chapter, we discuss the types of ncRNAs produced by the different herpesvirus families during infection, some of the cellular ncRNAs manipulated by these viruses, and the overall contributions of ncRNAs to the viral life cycle, influence on the host environment, and pathogenesis.


Asunto(s)
Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/virología , Herpesviridae/genética , Herpesviridae/fisiología , Interacciones Huésped-Patógeno/genética , ARN no Traducido/genética , Herpesviridae/patogenicidad , Humanos , ARN Viral/genética
9.
PLoS Pathog ; 12(11): e1006014, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27829026

RESUMEN

Cytomegaloviruses (CMV) are highly species-specific due to millennia of co-evolution and adaptation to their host, with no successful experimental cross-species infection in primates reported to date. Accordingly, full genome phylogenetic analysis of multiple new CMV field isolates derived from two closely related nonhuman primate species, Indian-origin rhesus macaques (RM) and Mauritian-origin cynomolgus macaques (MCM), revealed distinct and tight lineage clustering according to the species of origin, with MCM CMV isolates mirroring the limited genetic diversity of their primate host that underwent a population bottleneck 400 years ago. Despite the ability of Rhesus CMV (RhCMV) laboratory strain 68-1 to replicate efficiently in MCM fibroblasts and potently inhibit antigen presentation to MCM T cells in vitro, RhCMV 68-1 failed to productively infect MCM in vivo, even in the absence of host CD8+ T and NK cells. In contrast, RhCMV clone 68-1.2, genetically repaired to express the homologues of the HCMV anti-apoptosis gene UL36 and epithelial cell tropism genes UL128 and UL130 absent in 68-1, efficiently infected MCM as evidenced by the induction of transgene-specific T cells and virus shedding. Recombinant variants of RhCMV 68-1 and 68-1.2 revealed that expression of either UL36 or UL128 together with UL130 enabled productive MCM infection, indicating that multiple layers of cross-species restriction operate even between closely related hosts. Cumulatively, these results implicate cell tropism and evasion of apoptosis as critical determinants of CMV transmission across primate species barriers, and extend the macaque model of human CMV infection and immunology to MCM, a nonhuman primate species with uniquely simplified host immunogenetics.


Asunto(s)
Infecciones por Citomegalovirus/transmisión , Citomegalovirus/genética , Modelos Animales de Enfermedad , Macaca fascicularis/virología , Macaca mulatta/virología , Animales , Infecciones por Citomegalovirus/genética , ADN Viral/análisis , ADN Viral/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Filogenia , Especificidad de la Especie
10.
Rheumatol Immunol Res ; 4(2): 90-97, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37818347

RESUMEN

Objective: Synovial fibroblasts in patients with rheumatoid arthritis (RA) contribute substantially to the perpetuation of synovitis and invasion to cartilage and bone, and are potential therapeutic targets. Fibroblast activation protein (FAP) is highly expressed by RA synovial fibroblasts and the expression is relatively specific. We tested whether FAP can serve as a molecular target to modulate synovial fibroblasts for therapy in experimental arthritis. Methods: mRNA encoding consensus FAP (cFAP) was encapsulated in lipid nanoparticles (LNP) and was injected intramuscularly as vaccine prior to induction of collagen-induced arthritis (CIA) and collagen antibody induced arthritis (CAIA) in mice. Development of CIA and CAIA was assessed clinically and by histology. Results: cFAP mRNA-LNP vaccine provoked immune response to cFAP and mouse FAP (mFAP); prevented onset of CIA in 40% of mice and significantly reduced the severity of arthritis. In CAIA, cFAP mRNA-LNP did not prevent onset of arthritis but significantly reduced the severity of arthritis. Conclusion: cFAP mRNA-LNP vaccine was able to provoke immune response to mFAP and suppress inflammatory arthritis.

11.
mBio ; 13(1): e0172421, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35012351

RESUMEN

Human cytomegalovirus (HCMV) is a highly prevalent beta-herpesvirus and a significant cause of morbidity and mortality following hematopoietic and solid organ transplant, as well as the leading viral cause of congenital abnormalities. A key feature of the pathogenesis of HCMV is the ability of the virus to establish a latent infection in hematopoietic progenitor and myeloid lineage cells. The study of HCMV latency has been hampered by difficulties in obtaining and culturing primary cells, as well as an inability to quantitatively measure reactivating virus, but recent advances in both in vitro and in vivo models of HCMV latency and reactivation have led to a greater understanding of the interplay between host and virus. Key differences in established model systems have also led to controversy surrounding the role of viral gene products in latency establishment, maintenance, and reactivation. This review will discuss the details and challenges of various models including hematopoietic progenitor cells, monocytes, cell lines, and humanized mice. We highlight the utility and functional differences between these models and the necessary experimental design required to define latency and reactivation, which will help to generate a more complete picture of HCMV infection of myeloid-lineage cells.


Asunto(s)
Infecciones por Citomegalovirus , Citomegalovirus , Humanos , Animales , Ratones , Citomegalovirus/genética , Latencia del Virus/genética , Línea Celular , Células Madre Hematopoyéticas , Activación Viral/genética
12.
Sci Immunol ; 7(72): eabn9301, 2022 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-35714200

RESUMEN

The strain 68-1 rhesus cytomegalovirus (RhCMV)-based vaccine for simian immunodeficiency virus (SIV) can stringently protect rhesus macaques (RMs) from SIV challenge by arresting viral replication early in primary infection. This vaccine elicits unconventional SIV-specific CD8+ T cells that recognize epitopes presented by major histocompatibility complex (MHC)-II and MHC-E instead of MHC-Ia. Although RhCMV/SIV vaccines based on strains that only elicit MHC-II- and/or MHC-Ia-restricted CD8+ T cells do not protect against SIV, it remains unclear whether MHC-E-restricted T cells are directly responsible for protection and whether these responses can be separated from the MHC-II-restricted component. Using host microRNA (miR)-mediated vector tropism restriction, we show that the priming of MHC-II and MHC-E epitope-targeted responses depended on vector infection of different nonoverlapping cell types in RMs. Selective inhibition of RhCMV infection in myeloid cells with miR-142-mediated tropism restriction eliminated MHC-E epitope-targeted CD8+ T cell priming, yielding an exclusively MHC-II epitope-targeted response. Inhibition with the endothelial cell-selective miR-126 eliminated MHC-II epitope-targeted CD8+ T cell priming, yielding an exclusively MHC-E epitope-targeted response. Dual miR-142 + miR-126-mediated tropism restriction reverted CD8+ T cell responses back to conventional MHC-Ia epitope targeting. Although the magnitude and differentiation state of these CD8+ T cell responses were generally similar, only the vectors programmed to elicit MHC-E-restricted CD8+ T cell responses provided protection against SIV challenge, directly demonstrating the essential role of these responses in RhCMV/SIV vaccine efficacy.


Asunto(s)
Vacunas contra Citomegalovirus , MicroARNs , Vacunas contra el SIDAS , Síndrome de Inmunodeficiencia Adquirida del Simio , Virus de la Inmunodeficiencia de los Simios , Animales , Linfocitos T CD8-positivos , Citomegalovirus/genética , Epítopos , Macaca mulatta , Complejo Mayor de Histocompatibilidad , Células Mieloides , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Virus de la Inmunodeficiencia de los Simios/genética , Tropismo , Eficacia de las Vacunas
13.
J Virol ; 84(3): 1366-75, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19939931

RESUMEN

The herpes simplex virus (HSV) genome rapidly becomes associated with histones after injection into the host cell nucleus. The viral proteins ICP0 and VP16 are required for efficient viral gene expression and have been implicated in reducing the levels of underacetylated histones on the viral genome, raising the possibility that high levels of underacetylated histones inhibit viral gene expression. The U2OS osteosarcoma cell line is permissive for replication of ICP0 and VP16 mutants and appears to lack an innate antiviral repression mechanism present in other cell types. We therefore used chromatin immunoprecipitation to determine whether U2OS cells are competent to load histones onto HSV DNA and, if so, whether ICP0 and/or VP16 are required to reduce histone occupancy and enhance acetylation in this cell type. High levels of underacetylated histone H3 accumulated at several locations on the viral genome in the absence of VP16 activation function; in contrast, an ICP0 mutant displayed markedly reduced histone levels and enhanced acetylation, similar to wild-type HSV. These results demonstrate that U2OS cells are competent to load underacetylated histones onto HSV DNA and uncover an unexpected role for VP16 in modulating chromatin structure at viral early and late loci. One interpretation of these findings is that ICP0 and VP16 affect viral chromatin structure through separate pathways, and the pathway targeted by ICP0 is defective in U2OS cells. We also show that HSV infection results in decreased histone levels on some actively transcribed genes within the cellular genome, demonstrating that viral infection alters cellular chromatin structure.


Asunto(s)
Proteína Vmw65 de Virus del Herpes Simple/fisiología , Histonas/metabolismo , Proteínas Inmediatas-Precoces/fisiología , Osteosarcoma/virología , Ubiquitina-Proteína Ligasas/fisiología , Acetilación , Secuencia de Bases , Línea Celular Tumoral , Cartilla de ADN , Perfilación de la Expresión Génica , Herpesvirus Humano 1/genética , Humanos , Osteosarcoma/patología
14.
Pathogens ; 10(2)2021 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-33668486

RESUMEN

Human cytomegalovirus (HCMV) encodes 22 mature microRNAs (miRNAs), which regulate a myriad of cellular processes, including vesicular trafficking, cell cycle progression, apoptosis, and immune evasion, as well as viral gene expression. Recent evidence points to a critical role for HCMV miRNAs in mediating latency in CD34+ hematopoietic progenitor cells through modulation of cellular signaling pathways, including attenuation of TGFß and EGFR signaling. Moreover, HCMV miRNAs can act in concert with, or in opposition to, viral proteins in regulating host cell functions. Here, we comprehensively review the studies of HCMV miRNAs in the context of latency and highlight the novel processes that are manipulated by the virus using these small non-coding RNAs.

15.
mSphere ; 6(1)2021 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-33408225

RESUMEN

Human cytomegalovirus (HCMV) infection of myeloid lineage cells, such as CD34+ hematopoietic progenitor cells (HPCs) or monocytes, results in the upregulation of antiapoptotic cellular proteins that protect the newly infected cells from programmed cell death. The mechanisms used by HCMV to regulate proapoptotic cellular proteins upon infection of CD34+ HPCs have not been fully explored. Here, we show that HCMV utilizes pUL7, a secreted protein that signals through the FLT3 receptor, and miR-US5-1 and miR-UL112-3p to reduce the abundance and activity of the proapoptotic transcription factor FOXO3a at early times after infection of CD34+ HPCs. Regulation of FOXO3a by pUL7, miR-US5-1, and miR-UL112 results in reduced expression of the proapoptotic BCL2L11 transcript and protection of CD34+ HPCs from virus-induced apoptosis. These data highlight the importance of both viral proteins and microRNAs (miRNAs) in protecting CD34+ HPCs from apoptosis at early times postinfection, allowing for the establishment of latency and maintenance of viral genome-containing cells.IMPORTANCE Human cytomegalovirus (HCMV) causes serious disease in immunocompromised individuals and is a significant problem during transplantation. The virus can establish a latent infection in CD34+ hematopoietic progenitor cells (HPCs) and periodically reactivate to cause disease in the absence of an intact immune system. What viral gene products are required for successful establishment of latency is still not fully understood. Here, we show that both a viral protein and viral miRNAs are required to prevent apoptosis after infection of CD34+ HPCs. HCMV pUL7 and miRNAs miR-US5-1 and miR-UL112-3p act to limit the expression and activation of the transcription factor FOXO3a, which in turn reduces expression of proapoptotic gene BCL2L11 and prevents virus-induced apoptosis in CD34+ HPCs.


Asunto(s)
Antígenos CD34/genética , Apoptosis , Citomegalovirus/genética , Células Madre Hematopoyéticas/virología , MicroARNs/genética , Proteínas de la Matriz Viral/genética , Antígenos CD34/inmunología , Células Cultivadas , Fibroblastos/virología , Células HEK293 , Células Madre Hematopoyéticas/inmunología , Humanos , MicroARNs/clasificación
16.
mBio ; 12(2)2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33824207

RESUMEN

Human cytomegalovirus (HCMV) microRNAs play essential roles in latency and reactivation in CD34+ hematopoietic progenitor cells (HPCs) via regulation of viral and cellular gene expression. In the present study, we show that HCMV miR-US25-1 targets RhoA, a small GTPase required for CD34+ HPC self-renewal, proliferation, and hematopoiesis. Expression of miR-US25-1 impairs signaling through the nonmuscle myosin II light chain, which leads to a block in cytokinesis and an inhibition of proliferation. Moreover, infection with an HCMV mutant lacking miR-US25-1 resulted in increased proliferation of CD34+ HPCs and a decrease in the proportion of genome-containing cells at the end of latency culture. These observations provide a mechanism by which HCMV limits proliferation to maintain latent viral genomes in CD34+ HPCs.IMPORTANCE Each herpesvirus family establishes latency in a unique cell type. Since herpesvirus genomes are maintained as episomes, the virus needs to devise mechanisms to retain the latent genome during cell division. Alphaherpesviruses overcome this obstacle by infecting nondividing neurons, while gammaherpesviruses tether their genome to the host chromosome in dividing B cells. The betaherpesvirus human cytomegalovirus (HCMV) establishes latency in CD34+ hematopoietic progenitor cells (HPCs), but the mechanism used to maintain the viral genome is unknown. In this report, we demonstrate that HCMV miR-US25-1 downregulates expression of RhoA, a key cell cycle regulator, which results in inhibition of CD34+ HPC proliferation by blocking mitosis. Mutation of miR-US25-1 during viral infection results in enhanced cellular proliferation and a decreased frequency of genome-containing CD34+ HPCs. These results reveal a novel mechanism through which HCMV is able to regulate cell division to prevent viral genome loss during proliferation.


Asunto(s)
Antígenos CD34/genética , Proliferación Celular/genética , Citomegalovirus/genética , Genoma Viral , Células Madre Hematopoyéticas/fisiología , Interacciones Huésped-Patógeno , MicroARNs/genética , Latencia del Virus/genética , Proteína de Unión al GTP rhoA/genética , Antígenos CD34/inmunología , Antígenos CD34/metabolismo , Citomegalovirus/patogenicidad , Regulación hacia Abajo , Regulación de la Expresión Génica , Células HEK293 , Humanos , MicroARNs/metabolismo , Mitosis/genética , Transducción de Señal/genética , Proteína de Unión al GTP rhoA/inmunología
17.
Methods Mol Biol ; 2244: 301-342, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33555594

RESUMEN

microRNAs (miRNAs) are small noncoding RNAs that regulate gene expression at the posttranscriptional level by binding to sites within the 3' untranslated regions of messenger RNA (mRNA) transcripts. The discovery of this completely new mechanism of gene regulation necessitated the development of a variety of techniques to further characterize miRNAs, their expression, and function. In this chapter, we will discuss techniques currently used in the miRNA field to detect, express and inhibit miRNAs, as well as methods used to identify and validate their targets, specifically with respect to the miRNAs encoded by human cytomegalovirus.


Asunto(s)
Citomegalovirus/genética , Inmunoprecipitación/métodos , MicroARNs/análisis , Regiones no Traducidas 3'/genética , Northern Blotting/métodos , Expresión Génica/genética , Regulación Viral de la Expresión Génica/genética , Humanos , MicroARNs/genética , ARN Mensajero/genética
18.
Sci Immunol ; 6(57)2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33766849

RESUMEN

Simian immunodeficiency virus (SIV) insert-expressing, 68-1 rhesus cytomegalovirus (RhCMV/SIV) vectors elicit major histocompatibility complex E (MHC-E)- and MHC-II-restricted, SIV-specific CD8+ T cell responses, but the basis of these unconventional responses and their contribution to demonstrated vaccine efficacy against SIV challenge in the rhesus monkeys (RMs) have not been characterized. We show that these unconventional responses resulted from a chance genetic rearrangement in 68-1 RhCMV that abrogated the function of eight distinct immunomodulatory gene products encoded in two RhCMV genomic regions (Rh157.5/Rh157.4 and Rh158-161), revealing three patterns of unconventional response inhibition. Differential repair of these genes with either RhCMV-derived or orthologous human CMV (HCMV)-derived sequences (UL128/UL130; UL146/UL147) leads to either of two distinct CD8+ T cell response types-MHC-Ia-restricted only or a mix of MHC-II- and MHC-Ia-restricted CD8+ T cells. Response magnitude and functional differentiation are similar to RhCMV 68-1, but neither alternative response type mediated protection against SIV challenge. These findings implicate MHC-E-restricted CD8+ T cell responses as mediators of anti-SIV efficacy and indicate that translation of RhCMV/SIV vector efficacy to humans will likely require deletion of all genes that inhibit these responses from the HCMV/HIV vector.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Reprogramación Celular/inmunología , Infecciones por Citomegalovirus/veterinaria , Citomegalovirus/inmunología , Enfermedades de los Monos/prevención & control , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Vacunas Virales/inmunología , Animales , Antígenos Virales/genética , Antígenos Virales/inmunología , Linfocitos T CD8-positivos/metabolismo , Reprogramación Celular/genética , Ingeniería Genética/métodos , Vectores Genéticos/genética , Inmunogenicidad Vacunal , Memoria Inmunológica , Macaca mulatta , Enfermedades de los Monos/inmunología , Enfermedades de los Monos/virología , Sistemas de Lectura Abierta/genética , Sistemas de Lectura Abierta/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Virus de la Inmunodeficiencia de los Simios/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Eficacia de las Vacunas
19.
J Virol ; 83(17): 8976-9, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19535444

RESUMEN

Herpes simplex virus VP16 and ICP0 mutants replicate efficiently in U2OS osteosarcoma cells but are restricted in other cell types. We previously showed that the restrictive phenotype is dominant in a transient cell fusion assay, suggesting that U2OS cells lack an antiviral mechanism present in other cells. Recent data indicate that unscheduled membrane fusion events can activate the expression of interferon-stimulated genes (ISGs) in fibroblasts, raising the possibility that our earlier results were due to a fusion-induced antiviral state. However, we show here that the permissive phenotype is also extinguished following fusion with Vero cells in the absence of ISG induction.


Asunto(s)
Fusión Celular , Proteína Vmw65 de Virus del Herpes Simple/deficiencia , Proteínas Inmediatas-Precoces/deficiencia , Interferones/biosíntesis , Simplexvirus/inmunología , Simplexvirus/fisiología , Ubiquitina-Proteína Ligasas/deficiencia , Replicación Viral , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Humanos , Células Vero
20.
Mol Immunol ; 46(3): 393-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19038458

RESUMEN

The innate cellular response to virus particle entry in non-immune cells requires the transcriptional activity of interferon regulatory factor 3 (IRF-3), but not production of type I interferon (IFN). Here, we characterize the IFN-independent innate cellular response to virus-derived stimuli in Vero cells, a monkey kidney epithelial cell line deficient for IFN production. We provide evidence that Vero cells are deficient in their ability to mount an IRF-3-dependent, IFN-independent antiviral response against either incoming virus particles or polyinosinic:polycytidylic acid (pIC), a dsRNA mimetic. We further demonstrate that abundance of IRF-3 protein is a determinant in the pIC-mediated antiviral signalling pathway. These observations further characterize the permissive nature of Vero cells to viral infection, and highlight the crucial involvement of IRF-3 in the innate antiviral response.


Asunto(s)
Herpesvirus Humano 1/inmunología , Factor 3 Regulador del Interferón/inmunología , Interferones/biosíntesis , Virus Sendai/inmunología , Animales , Chlorocebus aethiops , Clonación Molecular , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 1/efectos de la radiación , Humanos , Poli I-C/farmacología , Virus Sendai/efectos de los fármacos , Virus Sendai/fisiología , Virus Sendai/efectos de la radiación , Análisis de Secuencia de ADN , Rayos Ultravioleta , Células Vero , Inactivación de Virus/efectos de los fármacos , Inactivación de Virus/efectos de la radiación , Replicación Viral/efectos de los fármacos , Replicación Viral/efectos de la radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA