Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
bioRxiv ; 2024 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-38328222

RESUMO

To establish a successful infection, herpes simplex virus-1 (HSV-1), a virus with high seropositivity in the human population, must undermine host innate and intrinsic immune defense mechanisms, including the stimulator of interferon genes (STING) pathway. Recently it was discovered that not only de novo produced intracellular 2'-3'cGAMP, but also extracellular 2'-3'cGAMP activates the STING pathway by being transported across the cell membrane via the folate transporter, SLC19A1, the first identified extracellular antiporter of this signaling molecule. We hypothesized that the import of exogenous 2'-3'cGAMP functions to establish an antiviral state like that seen with the paracrine antiviral activities of interferon. Further, to establish a successful infection, HSV-1 must undermine this induction of the STING pathway by inhibiting the biological functions of SLC19A1. Herein, we report that treatment of the monocytic cell line, THP-1 cells, epithelial cells (ARPE-19) and SH-SY5Y neuronal cell line with exogenous 2'-3'cGAMP induces interferon production and establishes an antiviral state. Using either pharmaceutical inhibition or genetic knockout of SLC19A1 blocks the 2'-3'cGAMP-induced antiviral state. Additionally, HSV-1 infection results in the reduction of SLC19A1 transcription, translation, and importantly, the rapid removal of SLC19A1 from the cell surface of infected cells. Our data indicate SLC19A1 functions as a newly identified antiviral mediator for extracellular 2'-3'cGAMP which is undermined by HSV-1. This work presents novel and important findings about how HSV-1 manipulates the host's immune environment for viral replication and discovers details about an antiviral mechanism which information could aid in the development of better antiviral drugs in the future. Importance: HSV-1 has evolved multiple mechanisms to neutralize of the host's innate and intrinsic defense pathways, such as the STING pathway. Here, we identified an antiviral response in which extracellular 2'-3'cGAMP triggers IFN production via its transporter SLC19A1. Moreover, we report that HSV-1 blocks the functions of this transporter thereby impeding the antiviral response, suggesting exogenous 2'-3'cGAMP can act as an immunomodulatory molecule in uninfected cells to activate the STING pathway, and priming an antiviral state, similar to that seen in interferon responses. The details of this mechanism highlight important details about HSV-1 infections. This work presents novel findings about how HSV-1 manipulates the host's immune environment for viral replication and reveals details about a novel antiviral mechanism. These findings expand our understanding of how viral infections undermine host responses and may help in the development of better broad based antiviral drugs in the future.

2.
J Clin Invest ; 133(12)2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37317966

RESUMO

Most drugs used to treat viral disease target a virus-coded product. They inhibit a single virus or virus family, and the pathogen can readily evolve resistance. Host-targeted antivirals can overcome these limitations. The broad-spectrum activity achieved by host targeting can be especially useful in combating emerging viruses and for treatment of diseases caused by multiple viral pathogens, such as opportunistic agents in immunosuppressed patients. We have developed a family of compounds that modulate sirtuin 2, an NAD+-dependent deacylase, and now report the properties of a member of that family, FLS-359. Biochemical and x-ray structural studies show that the drug binds to sirtuin 2 and allosterically inhibits its deacetylase activity. FLS-359 inhibits the growth of RNA and DNA viruses, including members of the coronavirus, orthomyxovirus, flavivirus, hepadnavirus, and herpesvirus families. FLS-359 acts at multiple levels to antagonize cytomegalovirus replication in fibroblasts, causing modest reductions in viral RNAs and DNA, together with a much greater reduction in infectious progeny, and it exhibits antiviral activity in humanized mouse models of infection. Our results highlight the potential of sirtuin 2 inhibitors as broad-spectrum antivirals and set the stage for further understanding of how host epigenetic mechanisms impact the growth and spread of viral pathogens.


Assuntos
Infecções por Coronavirus , Coronavirus , Animais , Camundongos , Antivirais/farmacologia , Sirtuína 2/genética , RNA Viral
3.
Viruses ; 14(6)2022 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-35746751

RESUMO

Human cytomegalovirus (CMV) is a ubiquitous pathogen that latently resides in hematopoietic cells. Latently infected individuals with dysfunctional immune systems often experience CMV reactivation, which can cause devastating disease and mortality. While factors dictating the balance between latency and reactivation are not completely understood, CMV US28 is required for maintaining latent infection, and viral mutants that alter US28 function result in a lytic-like, rather than latent, infection in hematopoietic cells. In turn, viral lytic factors alter the host cell, making it challenging to characterize the US28-specific changes in the cellular milieu. To circumvent this, we generated a temperature-sensitive TB40/E recombinant virus, TB40/EgfpC510G (tsC510G), into which we engineered an amino acid change at position 510 (C510G) of IE2, as previously described in the CMV Towne strain. Using tsC510G, we then deleted the US28 ORF, termed tsC510G-US28Δ. Consistent with previous findings, tsC510G-US28Δ fails to undergo latency in Kasumi-3 cells at the permissive temperature. However, parallel cultures maintained at the non-permissive temperature showed a significant reduction in infectious center frequency, as measured by limiting dilution assay. Thus, we generated a new US28 mutant virus for use as a tool to study US28-specific changes in latently infected hematopoietic cells in the absence of induced lytic replication.


Assuntos
Infecções por Citomegalovirus , Citomegalovirus , Citomegalovirus/fisiologia , Humanos , Temperatura , Proteínas Virais/genética , Proteínas Virais/metabolismo , Latência Viral , Replicação Viral
4.
Viruses ; 14(2)2022 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-35215840

RESUMO

Long non-coding RNA ß2.7 is the most highly transcribed viral gene during latent human cytomegalovirus (HCMV) infection. However, as yet, no function has ever been ascribed to ß2.7 during HCMV latency. Here we show that ß2.7 protects against apoptosis induced by high levels of reactive oxygen species (ROS) in infected monocytes, which routinely support latent HCMV infection. Monocytes infected with a wild-type (WT) virus, but not virus deleted for the ß2.7 gene (Δß2.7), are protected against mitochondrial stress and subsequent apoptosis. Protected monocytes display lower levels of ROS and additionally, stress-induced death in the absence of ß2.7 can be reversed by an antioxidant which reduces ROS levels. Furthermore, we show that infection with WT but not Δß2.7 virus results in strong upregulation of a cellular antioxidant enzyme, superoxide dismutase 2 (SOD2) in CD14+ monocytes. These observations identify a role for the ß2.7 viral transcript, the most abundantly expressed viral RNA during latency but for which no latency-associated function has ever been ascribed, and demonstrate a novel way in which HCMV protects infected monocytes from pro-death signals to optimise latent carriage.


Assuntos
Apoptose , Citomegalovirus/fisiologia , Monócitos/virologia , RNA Longo não Codificante/genética , RNA Viral/genética , Antioxidantes/metabolismo , Células Cultivadas , Citomegalovirus/genética , Humanos , Receptores de Lipopolissacarídeos/metabolismo , Mitocôndrias/metabolismo , Monócitos/metabolismo , Monócitos/patologia , Mutação , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Latência Viral/genética
5.
mBio ; 12(4): e0070021, 2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34425704

RESUMO

The necessity of viruses to modulate the innate immune response often dictates the outcome of viral infection. As such, viruses encode many factors that undermine these potent antiviral responses. A recent study by Bouvet et al. (M. Bouvet, S. Voigt, T. Tagawa, M. Albanese, et al., mBio 12:e03440-20, 2021, https://doi.org/10.1128/mBio.03440-20) revisits the impact of virus-encoded noncoding RNAs on key components of the interferon pathway and sheds light on how the extensive biological functions of Epstein-Barr virus (EBV) microRNAs (miRNAs) are on targeting both the induction and signaling cascades of interferon.


Assuntos
Infecções por Vírus Epstein-Barr , Herpesvirus Humano 4 , Antivirais , Herpesvirus Humano 4/genética , Humanos , Interferons , RNA Viral
6.
J Virol ; 94(17)2020 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-32581105

RESUMO

Human Cytomegalovirus (HCMV) is a ubiquitous pathogen that has coevolved with its host and, in doing so, is highly efficient in undermining antiviral responses that limit successful infections. As a result, HCMV infections are highly problematic in individuals with weakened or underdeveloped immune systems, including transplant recipients and newborns. Understanding how HCMV controls the microenvironment of an infected cell so as to favor productive replication is of critical importance. To this end, we took an unbiased proteomics approach to identify the highly reversible, stress-induced, posttranslational modification (PTM) protein S-nitrosylation on viral proteins to determine the biological impact on viral replication. We identified protein S-nitrosylation of 13 viral proteins during infection of highly permissive fibroblasts. One of these proteins, pp71, is critical for efficient viral replication, as it undermines host antiviral responses, including stimulator of interferon genes (STING) activation. By exploiting site-directed mutagenesis of the specific amino acids we identified in pp71 as protein S-nitrosylated, we found this pp71 PTM diminishes its ability to undermine antiviral responses induced by the STING pathway. Our results suggest a model in which protein S-nitrosylation may function as a host response to viral infection that limits viral spread.IMPORTANCE In order for a pathogen to establish a successful infection, it must undermine the host cell responses inhibitory to the pathogen. As such, herpesviruses encode multiple viral proteins that antagonize each host antiviral response, thereby allowing for efficient viral replication. Human Cytomegalovirus encodes several factors that limit host countermeasures to infection, including pp71. Herein, we identified a previously unreported posttranslational modification of pp71, protein S-nitrosylation. Using site-directed mutagenesis, we mutated the specific sites of this modification thereby blocking this pp71 posttranslational modification. In contexts where pp71 is not protein S-nitrosylated, host antiviral response was inhibited. The net result of this posttranslational modification is to render a viral protein with diminished abilities to block host responses to infection. This novel work supports a model in which protein S-nitrosylation may be an additional mechanism in which a cell inhibits a pathogen during the course of infection.


Assuntos
Antivirais/farmacologia , Citomegalovirus/efeitos dos fármacos , Proteína S/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular , Fibroblastos/virologia , Regulação Viral da Expressão Gênica , Humanos , Proteínas de Membrana/genética , Mutagênese Sítio-Dirigida , Proteínas Virais/efeitos dos fármacos , Proteínas Virais/genética , Replicação Viral
7.
Cell Rep ; 28(2): 434-448.e6, 2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31291579

RESUMO

Cellular SAMHD1 inhibits replication of many viruses by limiting intracellular deoxynucleoside triphosphate (dNTP) pools. We investigate the influence of SAMHD1 on human cytomegalovirus (HCMV). During HCMV infection, we observe SAMHD1 induction, accompanied by phosphorylation via viral kinase UL97. SAMHD1 depletion increases HCMV replication in permissive fibroblasts and conditionally permissive myeloid cells. We show this is due to enhanced gene expression from the major immediate-early (MIE) promoter and is independent of dNTP levels. SAMHD1 suppresses innate immune responses by inhibiting nuclear factor κB (NF-κB) activation. We show that SAMHD1 regulates the HCMV MIE promoter through NF-κB activation. Chromatin immunoprecipitation reveals increased RELA and RNA polymerase II on the HCMV MIE promoter in the absence of SAMHD1. Our studies reveal a mechanism of HCMV virus restriction by SAMHD1 and show how SAMHD1 deficiency activates an innate immune pathway that paradoxically results in increased viral replication through transcriptional activation of the HCMV MIE gene promoter.


Assuntos
Infecções por Citomegalovirus/virologia , Citomegalovirus/patogenicidade , NF-kappa B/metabolismo , Proteína 1 com Domínio SAM e Domínio HD/metabolismo , Humanos , Fosforilação , Regulação para Cima
8.
Viruses ; 10(11)2018 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-30405048

RESUMO

Human cytomegalovirus (HCMV) is a ubiquitous herpesvirus for which there is no vaccine or cure. This viral infection, once acquired, is life-long, residing latently in hematopoietic cells. However, latently infected individuals with weakened immune systems often undergo HCMV reactivation, which can cause serious complications in immunosuppressed and immunocompromised patients. Current anti-viral therapies target late stages of viral replication, and are often met with therapeutic resistance, necessitating the development of novel therapeutics. In this current study, we identified a naturally-occurring flavonoid compound, deguelin, which inhibits HCMV lytic replication. Our findings reveal that nanomolar concentrations of deguelin significantly suppress the production of the infectious virus. Further, we show that deguelin inhibits the lytic cycle during the phase of the replication cycle consistent with early (E) gene and protein expression. Importantly, our data reveal that deguelin inhibits replication of a ganciclovir-resistant strain of HCMV. Together, our findings identify a novel, naturally occurring compound that may prove useful in the treatment of HCMV replication.


Assuntos
Citomegalovirus/efeitos dos fármacos , Citomegalovirus/fisiologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/virologia , Flavonoides/farmacologia , Rotenona/análogos & derivados , Replicação Viral/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Farmacorresistência Viral , Flavonoides/química , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Rotenona/química , Rotenona/farmacologia , Internalização do Vírus/efeitos dos fármacos
9.
J Virol ; 92(21)2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30089702

RESUMO

Infections with human cytomegalovirus (HCMV) are highly prevalent in the general population as the virus has evolved the capacity to undergo distinct replication strategies resulting in lytic, persistent, and latent infections. During the latent life cycle, HCMV resides in subsets of cells within the hematopoietic cell compartment, including hematopoietic progenitor cells (HPCs) and peripheral blood monocytes. Since only a small fraction of these cell types harbor viral genomes during natural latency, identification and analysis of distinct changes mediated by viral infection are difficult to assess. In order to characterize latent infections of HPCs, we used an approach that involves complementation of deficiencies within the human pyrimidine salvage pathway, thus allowing for conversion of labeled uracil into rUTP. Here, we report the development of a recombinant HCMV that complements the defective human pyrimidine salvage pathway, allowing incorporation of thiol containing UTP into all RNA species that are synthesized within an infected cell. This virus grows to wild-type kinetics and can establish a latent infection within two distinct culture models of HCMV latency. Using this recombinant HCMV, we report the specific labeling of transcripts only within infected cells. These transcripts reveal a transcriptional landscape during HCMV latency that is distinct from uninfected cells. The utility of this labeling system allows for the identification of distinct changes within host transcripts and will shed light on characterizing how HCMV establishes and maintains latency.IMPORTANCE HCMV is a significant pathogen that accounts for a substantial amount of complications within the immunosuppressed and immunocompromised. Of particular significance is the capacity of HCMV to reactivate within solid tissue and bone marrow transplant recipients. While it is known that HCMV latency resides within a fraction of HPCs and monocytes, the exact subset of cells that harbor latent viral genomes during natural infections remain uncharacterized. The capacity to identify changes within the host transcriptome during latent infections is critical for developing approaches that therapeutically or physically eliminate latent viral genome containing cells and will represent a major breakthrough for reducing complications due to HCMV reactivation posttransplant. In this report, we describe the generation and use of a recombinant HCMV that allows specific and distinct labeling of RNA species that are produced within virally infected cells. This is a critical first step in identifying how HCMV affects the host cell during latency and more importantly, allows one to characterize cells that harbor latent HCMV.


Assuntos
Citomegalovirus/genética , Pentosiltransferases/genética , RNA Viral/genética , Coloração e Rotulagem/métodos , Tiouracila/análogos & derivados , Uracila/química , Células Cultivadas , Citomegalovirus/enzimologia , Infecções por Citomegalovirus , Humanos , Tiouracila/química , Latência Viral/genética
10.
Dev Cell ; 45(1): 83-100.e7, 2018 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-29634939

RESUMO

Human cytomegalovirus (HCMV), a leading cause of congenital birth defects, forms an unusual cytoplasmic virion maturation site termed the "assembly compartment" (AC). Here, we show that the AC also acts as a microtubule-organizing center (MTOC) wherein centrosome activity is suppressed and Golgi-based microtubule (MT) nucleation is enhanced. This involved viral manipulation of discrete functions of MT plus-end-binding (EB) proteins. In particular, EB3, but not EB1 or EB2, was recruited to the AC and was required to nucleate MTs that were rapidly acetylated. EB3-regulated acetylated MTs were necessary for nuclear rotation prior to cell migration, maintenance of AC structure, and optimal virus replication. Independently, a myristoylated peptide that blocked EB3-mediated enrichment of MT regulatory proteins at Golgi regions of the AC also suppressed acetylated MT formation, nuclear rotation, and infection. Thus, HCMV offers new insights into the regulation and functions of Golgi-derived MTs and the therapeutic potential of targeting EB3.


Assuntos
Núcleo Celular/fisiologia , Infecções por Citomegalovirus/virologia , Complexo de Golgi/virologia , Proteínas Associadas aos Microtúbulos/metabolismo , Centro Organizador dos Microtúbulos/fisiologia , Montagem de Vírus/fisiologia , Movimento Celular , Núcleo Celular/virologia , Células Cultivadas , Citomegalovirus/genética , Citomegalovirus/isolamento & purificação , Citomegalovirus/patogenicidade , Complexo de Golgi/fisiologia , Humanos , Proteínas Associadas aos Microtúbulos/genética , Centro Organizador dos Microtúbulos/virologia
11.
PLoS Pathog ; 13(11): e1006652, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29121116
12.
J Virol ; 90(8): 4249-4253, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26865717

RESUMO

The successful colonization of the majority of the population by human cytomegalovirus is a direct result of the virus's ability to establish and, more specifically, reactivate from latency. The underlying cellular factors involved in viral reactivation remain unknown. Here, we show that the host complexfacilitateschromatintranscription (FACT) binds to the major immediate early promoter (MIEP) and that inhibition of this complex reduces MIEP transactivation, thus inhibiting viral reactivation.


Assuntos
Citomegalovirus/fisiologia , Genes Precoces , Proteínas Virais/antagonistas & inibidores , Replicação Viral , Citomegalovirus/genética , Fibroblastos , Regulação Viral da Expressão Gênica , Humanos , Modelos Biológicos , Regiões Promotoras Genéticas , Transcrição Gênica , Proteínas Virais/metabolismo , Latência Viral , Liberação de Vírus
13.
J Virol ; 89(5): 2615-27, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25520507

RESUMO

UNLABELLED: Human herpesvirus 6A (HHV-6A), a member of the betaherpesvirus family, is associated with several human diseases. Like all herpesviruses, HHV-6A establishes a lifelong, latent infection in its host. Reactivation of HHV-6A is frequent within the immunosuppressed and immunocompromised populations and results in lytic viral replication within multiple organs, often leading to severe disease. MicroRNAs (miRNAs) are key regulators of multiple cellular processes that regulate the translation of specific transcripts. miRNAs carried by herpesviruses play important roles in modulating the host cell, thereby facilitating a suitable environment for productive viral infection and/or latency. Currently, there are approximately 150 known human herpesvirus-encoded miRNAs, although an miRNA(s) encoded by HHV-6A has yet to be reported. We hypothesized that HHV-6A, like other members of the human herpesvirus family, encodes miRNAs, which function to promote viral infection. We utilized deep sequencing of small RNA species isolated from cells harboring HHV-6A to identify five novel small noncoding RNA species that originate from the viral genome, one of which has the characteristics of a viral miRNA. These RNAs are expressed during productive infection by either bacterial artificial chromosome (BAC)-derived virus in Jjhan cells or wild-type HHV-6A strain U1102 virus in HSB2 cells and are associated with the RNA induced silencing complex (RISC) machinery. Growth analyses of mutant viruses that lack each individual miRNA revealed that a viral miRNA candidate (miR-U86) targets the HHV-6A IE gene U86, thereby regulating lytic replication. The identification and biological characterization of this HHV-6A-specific miRNA is the first step to defining how the virus regulates its life cycle. IMPORTANCE: A majority of the human population is infected with human herpesvirus 6A (HHV-6A), a betaherpesvirus family member. Infections usually occur in young children, and upon resolution, the virus remains in a latent state within the host. Importantly, during times of weakened immune responses, the virus can reactivate and is correlated with significant disease states. Viruses encode many different types of factors that both undermine the host antiviral response and regulate viral replication, including small RNA species called microRNAs (miRNAs). Here we report that HHV-6A encodes at least one miRNA, which we named miR-U86. We have characterized the requirement of this viral miRNA and its impact on the viral life cycle and found that it functions to regulate a viral protein important for efficient viral replication. Our data suggest that viral miRNAs are important for HHV-6A and that they may serve as an important therapeutic target to inhibit the virus.


Assuntos
Herpesvirus Humano 6/fisiologia , MicroRNAs/metabolismo , RNA Viral/metabolismo , Replicação Viral , Linhagem Celular , Perfilação da Expressão Gênica , Herpesvirus Humano 6/genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , MicroRNAs/genética , RNA Viral/genética
14.
Curr Opin Virol ; 9: 188-93, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25437230

RESUMO

Recent technological advances have led to an explosion in the system-wide profiling of biological processes in the study of herpesvirus biology, herein referred to as '-omics'. In many cases these approaches have revealed novel virus-induced changes to host cell biology that can be targeted with new antiviral therapeutics. Despite these successes, -omics approaches are not widely applied in the study of roseoloviruses. Here we describe examples of how -omics studies have shaped our understanding of herpesvirus biology, and discuss how these approaches might be used to identify host and viral factors that mediate roseolovirus pathogenesis.


Assuntos
Interações Hospedeiro-Patógeno , Roseolovirus/genética , Roseolovirus/fisiologia , Biologia de Sistemas/métodos , Perfilação da Expressão Gênica/métodos , Genômica/métodos , Humanos , Metabolômica/métodos , Proteômica/métodos , Roseolovirus/química , Biologia de Sistemas/tendências , Virologia/métodos , Virologia/tendências
15.
J Virol ; 88(10): 5524-32, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24599990

RESUMO

UNLABELLED: Reactivation of human cytomegalovirus (HCMV) is a significant cause of disease and death in immunocompromised patients, underscoring the need to understand how latency is controlled. Here we demonstrate that HCMV has evolved to utilize cellular microRNAs (miRNAs) in cells that promote latency to regulate expression of a viral protein critical for viral reactivation. Our data reveal that hsa-miR-200 miRNA family members target the UL122 (immediate early protein 2) 3' untranslated region, resulting in repression of this viral protein. Utilizing recombinant viruses that mutate the miRNA-binding site compared to the sequence of the wild-type virus results in lytic rather than latent infections in ex vivo infections of primary CD34+ cells. Cells permissive for lytic replication demonstrate low levels of these miRNAs. We propose that cellular miRNA regulation of HCMV is critical for maintenance of viral latency. IMPORTANCE: Human cytomegalovirus (HCMV) is a herpesvirus that infects a majority of the population. Once acquired, individuals harbor the virus for life, where the virus remains, for the most part, in a quiet or latent state. Under weakened immune conditions, the virus can reactivate, which can cause severe disease and often death. We have found that members of a family of small RNAs, termed microRNAs, encoded by human myeloid progenitor cells are capable of repressing a key viral protein, thus enabling the virus to ensure a quiet/latent state. As these progenitor cells mature further down the myeloid lineage toward cells that support active viral replication, the levels of these microRNAs decrease. Together, our data suggest that host cell microRNA regulation of HCMV is important for the quiet/latent state of this pathogen.


Assuntos
Citomegalovirus/fisiologia , Regulação Viral da Expressão Gênica , Interações Hospedeiro-Patógeno , Proteínas Imediatamente Precoces/metabolismo , MicroRNAs/metabolismo , Biossíntese de Proteínas , Transativadores/metabolismo , Latência Viral , Linhagem Celular , Análise Mutacional de DNA , Humanos , Proteínas Imediatamente Precoces/genética , MicroRNAs/genética , Transativadores/genética
16.
PLoS Genet ; 9(10): e1003879, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24204294

RESUMO

Retrotransposon sequences are positioned throughout the genome of almost every eukaryote that has been sequenced. As mobilization of these elements can have detrimental effects on the transcriptional regulation and stability of an organism's genome, most organisms have evolved mechanisms to repress their movement. Here, we identify a novel role for the Drosophila melanogaster Condensin II subunit, dCAP-D3 in preventing the mobilization of retrotransposons located in somatic cell euchromatin. dCAP-D3 regulates transcription of euchromatic gene clusters which contain or are proximal to retrotransposon sequence. ChIP experiments demonstrate that dCAP-D3 binds to these loci and is important for maintaining a repressed chromatin structure within the boundaries of the retrotransposon and for repressing retrotransposon transcription. We show that dCAP-D3 prevents accumulation of double stranded DNA breaks within retrotransposon sequence, and decreased dCAP-D3 levels leads to a precise loss of retrotransposon sequence at some dCAP-D3 regulated gene clusters and a gain of sequence elsewhere in the genome. Homologous chromosomes exhibit high levels of pairing in Drosophila somatic cells, and our FISH analyses demonstrate that retrotransposon-containing euchromatic loci are regions which are actually less paired than euchromatic regions devoid of retrotransposon sequences. Decreased dCAP-D3 expression increases pairing of homologous retrotransposon-containing loci in tissue culture cells. We propose that the combined effects of dCAP-D3 deficiency on double strand break levels, chromatin structure, transcription and pairing at retrotransposon-containing loci may lead to 1) higher levels of homologous recombination between repeats flanking retrotransposons in dCAP-D3 deficient cells and 2) increased retrotransposition. These findings identify a novel role for the anti-pairing activities of dCAP-D3/Condensin II and uncover a new way in which dCAP-D3/Condensin II influences local chromatin structure to help maintain genome stability.


Assuntos
Adenosina Trifosfatases/genética , Cromossomos/genética , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Complexos Multiproteicos/genética , Retroelementos/genética , Adenosina Trifosfatases/biossíntese , Animais , Estruturas Cromossômicas/genética , Estruturas Cromossômicas/metabolismo , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA/biossíntese , Diploide , Proteínas de Drosophila/biossíntese , Drosophila melanogaster , Eucromatina/genética , Regulação da Expressão Gênica , Instabilidade Genômica , Complexos Multiproteicos/biossíntese , Análise de Sequência com Séries de Oligonucleotídeos
17.
J Virol ; 87(22): 12020-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24006442

RESUMO

Although serious human diseases have been correlated with human herpesvirus 6A (HHV-6A) and HHV-6B, the lack of animal models has prevented studies which would more definitively link these viral infections to disease. HHV-6A and HHV-6B have recently been classified as two distinct viruses, and in this study we focused specifically on developing an in vivo model for HHV-6A. Here we show that Rag2⁻/⁻γc⁻/⁻ mice humanized with cord blood-derived human hematopoietic stem cells produce human T cells that express the major HHV-6A receptor, CD46. Both cell-associated and cell-free viral transmission of HHV-6A into the peritoneal cavity resulted in detectable viral DNA in at least one of the samples (blood, bone marrow, etc.) analyzed from nearly all engrafted mice. Organs and cells positive for HHV-6A DNA were the plasma and cellular blood fractions, bone marrow, lymph node, and thymic samples; control mice had undetectable viral DNA. We also noted viral pathogenic effects on certain T cell populations. Specific thymocyte populations, including CD3⁻ CD4⁺ CD8⁻ and CD3⁺ CD4⁻ cells, were significantly modified in humanized mice infected by cell-associated transmission. In addition, we detected significantly increased proportions of CD4⁺ CD8⁺ cells in the blood of animals infected by cell-free transmission. These findings provide additional evidence that HHV-6A may play a role in human immunodeficiencies. These results indicate that humanized mice can be used to study HHV-6A in vivo infection and replication as well as aspects of viral pathogenesis.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Células-Tronco Hematopoéticas/patologia , Herpesvirus Humano 6/patogenicidade , Infecções por Roseolovirus/transmissão , Replicação Viral , Animais , Medula Óssea/imunologia , Medula Óssea/patologia , Medula Óssea/virologia , Células Cultivadas , DNA Viral/genética , Sangue Fetal/imunologia , Sangue Fetal/virologia , Citometria de Fluxo , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/virologia , Humanos , Linfonodos/imunologia , Linfonodos/patologia , Linfonodos/virologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Roseolovirus/imunologia , Infecções por Roseolovirus/virologia , Baço/imunologia , Baço/patologia , Baço/virologia , Timo/imunologia , Timo/patologia , Timo/virologia
18.
J Virol ; 86(18): 9854-65, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22761372

RESUMO

Human cytomegalovirus (HCMV) is a herpesvirus that establishes a lifelong, latent infection within a host. At times when the immune system is compromised, the virus undergoes a lytic reactivation producing infectious progeny. The identification and understanding of the biological mechanisms underlying HCMV latency and reactivation are not completely defined. To this end, we have developed a tractable in vitro model system to investigate these phases of viral infection using a clonal population of myeloid progenitor cells (Kasumi-3 cells). Infection of these cells results in maintenance of the viral genome with restricted viral RNA expression that is reversed with the addition of the phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA, also known as PMA). Additionally, a latent viral transcript (LUNA) is expressed at times where viral lytic transcription is suppressed. Infected Kasumi-3 cells initiate production of infectious virus following TPA treatment, which requires cell-to-cell contact for efficient transfer of virus to other cell types. Importantly, lytically infected fibroblast, endothelial, or epithelial cells can transfer virus to Kasumi-3 cells, which fail to initiate lytic replication until stimulated with TPA. Finally, inflammatory cytokines, in addition to the pharmacological agent TPA, are sufficient for transcription of immediate-early (IE) genes following latent infection. Taken together, our findings argue that the Kasumi-3 cell line is a tractable in vitro model system with which to study HCMV latency and reactivation.


Assuntos
Citomegalovirus/fisiologia , Citomegalovirus/patogenicidade , Células Progenitoras Mieloides/virologia , Sequência de Bases , Linhagem Celular , Citocinas/fisiologia , Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , DNA Viral/genética , Expressão Gênica/efeitos dos fármacos , Genes Precoces , Genoma Viral , Humanos , Mediadores da Inflamação/fisiologia , Modelos Biológicos , Células Progenitoras Mieloides/efeitos dos fármacos , Células Progenitoras Mieloides/fisiologia , Acetato de Tetradecanoilforbol/farmacologia , Ativação Viral/genética , Ativação Viral/fisiologia , Latência Viral/genética , Latência Viral/fisiologia , Replicação Viral/genética , Replicação Viral/fisiologia
19.
J Virol ; 80(14): 6873-82, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16809293

RESUMO

Murine cytomegalovirus (MCMV) and human CMV (HCMV) share many features making the mouse system a potential small-animal model for HCMV. Although the genomic DNA sequence and the predicted open reading frames (ORFs) of MCMV have been determined, experimental evidence that the ORFs are actually transcribed has been lacking. We developed an MCMV global-DNA microarray that includes all previously predicted ORFs and 14 potential ones. A total of 172 ORFs were confirmed to be transcribed, including 7 newly discovered ORFs not previously predicted. No gene products from 10 previously predicted ORFs were detected by either DNA microarray analysis or reverse transcriptase PCR in MCMV-infected mouse fibroblasts, although 2 of those were expressed in a macrophage cell line, suggesting that potential gene products from these open reading frames are silenced in fibroblasts and required in macrophages. Immunohistochemical localization of the six newly described ORF products and three recently identified ones in cells transfected with the respective construct revealed four of the products in the nucleus and five in mitochondria. Analysis of two ORFs using site-directed mutagenesis showed that deletion of one of the mitochondrion-localized gene products led to significantly decreased replication in fibroblasts.


Assuntos
Genes Virais/genética , Muromegalovirus/genética , Fases de Leitura Aberta/genética , Transcrição Gênica/genética , Animais , Núcleo Celular/genética , Núcleo Celular/imunologia , Núcleo Celular/virologia , Citomegalovirus/genética , Citomegalovirus/imunologia , Perfilação da Expressão Gênica , Inativação Gênica , Genes Virais/imunologia , Humanos , Imuno-Histoquímica , Camundongos , Mitocôndrias/genética , Mitocôndrias/imunologia , Mitocôndrias/virologia , Muromegalovirus/imunologia , Células NIH 3T3 , Análise de Sequência com Séries de Oligonucleotídeos , Fases de Leitura Aberta/imunologia , Transcrição Gênica/imunologia , Replicação Viral/genética , Replicação Viral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA