Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
mBio ; 14(1): e0354222, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36692302

RESUMO

Transcription of herpes simplex virus 1 (HSV-1) immediate early (IE) genes is controlled at multiple levels by the cellular transcriptional coactivator, HCF-1. HCF-1 is complexed with epigenetic factors that prevent silencing of the viral genome upon infection, transcription factors that drive initiation of IE gene expression, and transcription elongation factors required to circumvent RNAPII pausing at IE genes and promote productive IE mRNA synthesis. Significantly, the coactivator is also implicated in the control of viral reactivation from latency in sensory neurons based on studies that demonstrate that HCF-1-associated epigenetic and transcriptional elongation complexes are critical to initiate IE expression and viral reactivation. Here, an HCF-1 conditional knockout mouse model (HCF-1cKO) was derived to probe the role and significance of HCF-1 in the regulation of HSV-1 latency/reactivation in vivo. Upon deletion of HCF-1 in sensory neurons, there is a striking reduction in the number of latently infected neurons that initiate viral reactivation. Importantly, this correlated with a defect in the removal of repressive chromatin associated with latent viral genomes. These data demonstrate that HCF-1 is a critical regulatory factor that governs the initiation of HSV reactivation, in part, by promoting the transition of latent viral genomes from a repressed heterochromatic state. IMPORTANCE Herpes simplex virus is responsible for a substantial worldwide disease burden. An initial infection leads to the establishment of a lifelong persistent infection in sensory neurons. Periodic reactivation can result in recurrent oral and genital lesions to more significant ocular disease. Despite the significance of this pathogen, many of the regulatory factors and molecular mechanisms that govern the viral latency-reactivation cycles have yet to be elucidated. Initiation of both lytic infection and reactivation are dependent on the expression of the viral immediate early genes. In vivo deletion of a central component of the IE regulatory paradigm, the cellular transcriptional coactivator HCF-1, reduces the epigenetic transition of latent viral genomes, thus suppressing HSV reactivation. These observations define HCF-1 as a critical regulator that controls the initiation of HSV reactivation from latency in vivo and contribute to understanding of the molecular mechanisms that govern viral reactivation.


Assuntos
Herpes Simples , Herpesvirus Humano 1 , Animais , Camundongos , Regulação Viral da Expressão Gênica , Herpesvirus Humano 1/fisiologia , Heterocromatina , Fatores de Transcrição/metabolismo , Transcrição Gênica , Latência Viral/fisiologia
2.
mBio ; 11(3)2020 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-32518191

RESUMO

Induction of herpes simplex virus (HSV) immediate early (IE) gene transcription promotes the initiation of lytic infection and reactivation from latency in sensory neurons. IE genes are transcribed by the cellular RNA polymerase II (RNAPII) and regulated by multiple transcription factors and coactivators. The HCF-1 cellular coactivator plays a central role in driving IE expression at multiple stages through interactions with transcription factors, chromatin modulation complexes, and transcription elongation components, including the active super elongation complex/P-TEFb (SEC-P-TEFb). Here, we demonstrate that the SEC occupies the promoters of HSV IE genes during the initiation of lytic infection and during reactivation from latency. Specific inhibitors of the SEC suppress viral IE expression and block the spread of HSV infection. Significantly, these inhibitors also block the initiation of viral reactivation from latency in sensory ganglia. The potent suppression of IE gene expression by SEC inhibitors indicates that transcriptional elongation represents a determining rate-limiting stage in HSV IE gene transcription and that the SEC plays a critical role in driving productive elongation during both phases of the viral life cycle. Most importantly, this supports the model that signal-mediated induction of SEC-P-TEFb levels can promote reactivation of a population of poised latent genomes.IMPORTANCE HSV infections can cause pathologies ranging from recurrent lesions to significant ocular disease. Initiation of lytic infection and reactivation from latency in sensory neurons are dependent on the induced expression of the viral immediate early genes. Transcription of these genes is controlled at multiple levels, including modulation of the chromatin state of the viral genome and appropriate recruitment of transcription factors and coactivators. Following initiation of transcription, IE genes are subject to a key regulatory stage in which transcriptional elongation rates are controlled by the activity of the super elongation complex. Inhibition of the SEC blocks both lytic infection and reactivation from latency in sensory neurons. In addition to providing insights into the mechanisms controlling viral infection and reactivation, inhibitors of critical components such as the SEC may represent novel antivirals.


Assuntos
Expressão Gênica , Genes Precoces , Herpesvirus Humano 1/genética , Proteínas Imediatamente Precoces/genética , Fatores de Elongação da Transcrição/antagonistas & inibidores , Latência Viral/genética , Animais , Antivirais/farmacologia , Linhagem Celular , Chlorocebus aethiops , Fibroblastos/virologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/fisiologia , Humanos , Pulmão/citologia , Fatores de Elongação da Transcrição/genética , Células Vero , Latência Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
3.
J Virol ; 92(17)2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29899098

RESUMO

Following productive infection, bovine herpesvirus 1 (BoHV-1) establishes latency in sensory neurons. As in other alphaherpesviruses, expression of BoHV-1 immediate early (IE) genes is regulated by an enhancer complex containing the viral IE activator VP16, the cellular transcription factor Oct-1, and transcriptional coactivator HCF-1, which is assembled on an IE enhancer core element (TAATGARAT). Expression of the IE transcription unit that encodes the viral IE activators bICP0 and bICP4 may also be induced by the activated glucocorticoid receptor (GR) via two glucocorticoid response elements (GREs) located upstream of the enhancer core. Strikingly, lytic infection and reactivation from latency are consistently enhanced by glucocorticoid treatment in vivo As the coactivator HCF-1 is essential for IE gene expression of alphaherpesviruses and recruited by multiple transcription factors, we tested whether HCF-1 is required for glucocorticoid-induced IE gene expression. Depletion of HCF-1 reduced GR-mediated activation of the IE promoter in mouse neuroblastoma cells (Neuro-2A). More importantly, HCF-1-mediated GR activation of the promoter was dependent on the presence of GRE sites but independent of the TAATGARAT enhancer core element. HCF-1 was also recruited to the GRE region of a promoter lacking the enhancer core, consistent with a direct role of the coactivator in mediating GR-induced transcription. Similarly, during productive lytic infection, HCF-1 and GR occupied the IE region containing the GREs. These studies indicate HCF-1 is critical for GR activation of the viral IE genes and suggests that glucocorticoid induction of viral reactivation proceeds via an HCF-1-GR mechanism in the absence of the viral IE activator VP16.IMPORTANCE BoHV-1 transcription is rapidly activated during stress-induced reactivation from latency. The immediate early transcription unit 1 (IEtu1) promoter is regulated by the GR via two GREs. The IEtu1 promoter regulates expression of two viral transcriptional regulatory proteins, infected cell proteins 0 and 4 (bICP0 and bICP4), and thus must be stimulated during reactivation. This study demonstrates that activation of the IEtu1 promoter by the synthetic corticosteroid dexamethasone requires HCF-1. Interestingly, the GRE sites, but not the IE enhancer core element (TAATGARAT), were required for HCF-1-mediated GR promoter activation. The GR and HCF-1 were recruited to the IEtu1 promoter in transfected and infected cells. Collectively, these studies indicate that HCF-1 is critical for GR activation of the viral IE genes and suggest that an HCF-1-GR complex can stimulate the IEtu1 promoter in the absence of the viral IE activator VP16.


Assuntos
Regulação Viral da Expressão Gênica , Genes Precoces , Glucocorticoides/metabolismo , Herpesvirus Bovino 1/fisiologia , Fator C1 de Célula Hospedeira/metabolismo , Receptores de Glucocorticoides/metabolismo , Transcrição Gênica , Animais , Linhagem Celular , Técnicas de Silenciamento de Genes , Fator C1 de Célula Hospedeira/genética , Camundongos , Neurônios/virologia
4.
Cell Host Microbe ; 18(6): 649-58, 2015 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-26651941

RESUMO

Herpes simplex virus (HSV) reactivation from latent neuronal infection requires stimulation of lytic gene expression from promoters associated with repressive heterochromatin. Various neuronal stresses trigger reactivation, but how these stimuli activate silenced promoters remains unknown. We show that a neuronal pathway involving activation of c-Jun N-terminal kinase (JNK), common to many stress responses, is essential for initial HSV gene expression during reactivation. This JNK activation in neurons is mediated by dual leucine zipper kinase (DLK) and JNK-interacting protein 3 (JIP3), which direct JNK toward stress responses instead of other cellular functions. Surprisingly, JNK-mediated viral gene induction occurs independently of histone demethylases that remove repressive lysine modifications. Rather, JNK signaling results in a histone methyl/phospho switch on HSV lytic promoters, a mechanism permitting gene expression in the presence of repressive lysine methylation. JNK is present on viral promoters during reactivation, thereby linking a neuronal-specific stress pathway and HSV reactivation from latency.


Assuntos
Histonas/metabolismo , Neurônios/virologia , Processamento de Proteína Pós-Traducional , Simplexvirus/fisiologia , Ativação Viral , Animais , Células Cultivadas , Regulação da Expressão Gênica , Camundongos , Fosforilação , Regiões Promotoras Genéticas , Transdução de Sinais , Simplexvirus/genética , Estresse Fisiológico
5.
Sci Transl Med ; 6(265): 265ra169, 2014 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-25473037

RESUMO

Herpesviruses are highly prevalent and maintain lifelong latent reservoirs, thus posing challenges to the control of herpetic disease despite the availability of antiviral pharmaceuticals that target viral DNA replication. The initiation of herpes simplex virus infection and reactivation from latency is dependent on a transcriptional coactivator complex that contains two required histone demethylases, LSD1 (lysine-specific demethylase 1) and a member of the JMJD2 family (Jumonji C domain-containing protein 2). Inhibition of either of these enzymes results in heterochromatic suppression of the viral genome and blocks infection and reactivation in vitro. We demonstrate that viral infection can be epigenetically suppressed in three animal models of herpes simplex virus infection and disease. Treating animals with the monoamine oxidase inhibitor tranylcypromine to inhibit LSD1 suppressed viral lytic infection, subclinical shedding, and reactivation from latency in vivo. This phenotypic suppression was correlated with enhanced epigenetic suppression of the viral genome and suggests that, even during latency, the chromatin state of the virus is dynamic. Therefore, epi-pharmaceuticals may represent a promising approach to treat herpetic diseases.


Assuntos
Epigênese Genética , Infecções por Herpesviridae/metabolismo , Oxirredutases N-Desmetilantes/antagonistas & inibidores , Oxirredutases N-Desmetilantes/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Genoma Viral , Cobaias , Histona Desmetilases , Camundongos , Camundongos Endogâmicos BALB C , Inibidores da Monoaminoxidase/química , Fenótipo , Estrutura Terciária de Proteína , Coelhos , Recidiva , Tranilcipromina/química , Vagina/virologia , Ativação Viral , Latência Viral , Replicação Viral/efeitos dos fármacos , Eliminação de Partículas Virais
6.
Virology ; 449: 120-32, 2014 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-24418545

RESUMO

Classical inbred mice are extensively used for virus research. However, we recently found that some wild-derived inbred mouse strains are more susceptible than classical strains to monkeypox virus. Experiments described here indicated that the 50% lethal dose of vaccinia virus (VACV) and cowpox virus (CPXV) were two logs lower in wild-derived inbred CAST/Ei mice than classical inbred BALB/c mice, whereas there was little difference in the susceptibility of the mouse strains to herpes simplex virus. Live bioluminescence imaging was used to follow spread of pathogenic and attenuated VACV strains and CPXV virus from nasal passages to organs in the chest and abdomen of CAST/Ei mice. Luminescence increased first in the head and then simultaneously in the chest and abdomen in a dose-dependent manner. The spreading kinetics was more rapid with VACV than CPXV although the peak photon flux was similar. These data suggest advantages of CAST/Ei mice for orthopoxvirus studies.


Assuntos
Animais Selvagens/virologia , Vírus da Varíola Bovina/patogenicidade , Varíola Bovina/virologia , Modelos Animais de Doenças , Vaccinia virus/patogenicidade , Vacínia/virologia , Animais , Linhagem Celular , Chlorocebus aethiops , Vírus da Varíola Bovina/genética , Vírus da Varíola Bovina/fisiologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos/virologia , Vaccinia virus/genética , Vaccinia virus/fisiologia , Virulência , Imagem Corporal Total
7.
Viruses ; 5(5): 1272-91, 2013 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-23698399

RESUMO

Successful infection of herpes simplex virus is dependent upon chromatin modulation by the cellular coactivator host cell factor-1 (HCF-1). This review focuses on the multiple chromatin modulation components associated with HCF-1 and the chromatin-related dynamics mediated by this coactivator that lead to the initiation of herpes simplex virus (HSV) immediate early gene expression.


Assuntos
Cromatina/metabolismo , Fator C1 de Célula Hospedeira/metabolismo , Interações Hospedeiro-Patógeno , Simplexvirus/fisiologia , Replicação Viral , Regulação Viral da Expressão Gênica , Humanos
8.
mBio ; 4(1): e00558-12, 2013 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-23386436

RESUMO

Cellular processes requiring access to the DNA genome are regulated by an overlay of epigenetic modifications, including histone modification and chromatin remodeling. Similar to the cellular host, many nuclear DNA viruses that depend upon the host cell's transcriptional machinery are also subject to the regulatory impact of chromatin assembly and modification. Infection of cells with alphaherpesviruses (herpes simplex virus [HSV] and varicella-zoster virus [VZV]) results in the deposition of nucleosomes bearing repressive histone H3K9 methylation on the viral genome. This repressive state is modulated by the recruitment of a cellular coactivator complex containing the histone H3K9 demethylase LSD1 to the viral immediate-early (IE) gene promoters. Inhibition of the activity of this enzyme results in increased repressive chromatin assembly and suppression of viral gene expression during lytic infection as well as reactivation from latency in a mouse ganglion explant model. However, available small-molecule LSD1 inhibitors are not originally designed to inhibit LSD1, but rather monoamine oxidases (MAO) in general. Thus, their specificity for and potency to LSD1 is low. In this study, a novel specific LSD1 inhibitor was identified that potently repressed HSV IE gene expression, genome replication, and reactivation from latency. Importantly, the inhibitor also suppressed primary infection of HSV in vivo in a mouse model. Based on common control of a number of DNA viruses by epigenetic modulation, it was also demonstrated that this LSD1 inhibitor blocks initial gene expression of the human cytomegalovirus and adenovirus type 5. IMPORTANCE Epigenetic mechanisms, including histone modification and chromatin remodeling, play important regulatory roles in all cellular processes requiring access to the genome. These mechanisms are often altered in disease conditions, including various cancers, and thus represent novel targets for drugs. Similarly, many viral pathogens are regulated by an epigenetic overlay that determines the outcome of infection. Therefore, these epigenetic targets also represent novel antiviral targets. Here, a novel inhibitor was identified with high specificity and potency for the histone demethylase LSD1, a critical component of the herpes simplex virus (HSV) gene expression paradigm. This inhibitor was demonstrated to have potent antiviral potential in both cultured cells and animal models. Thus, in addition to clearly demonstrating the critical role of LSD1 in regulation of HSV infection, as well as other DNA viruses, the data extends the therapeutic potential of chromatin modulation inhibitors from the focused field of oncology to the arena of antiviral agents.


Assuntos
Antivirais/metabolismo , Histona Desmetilases/antagonistas & inibidores , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Simplexvirus/efeitos dos fármacos , Simplexvirus/fisiologia , Ativação Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Adenoviridae/efeitos dos fármacos , Adenoviridae/fisiologia , Animais , Linhagem Celular , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/fisiologia , Modelos Animais de Doenças , Feminino , Herpes Simples/tratamento farmacológico , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Simplexvirus/crescimento & desenvolvimento , Carga Viral
9.
Sci Transl Med ; 5(167): 167ra5, 2013 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-23303604

RESUMO

Chromatin and the chromatin modulation machinery not only provide a regulatory matrix for enabling cellular functions such as DNA replication and transcription but also regulate the infectious cycles of many DNA viruses. Elucidation of the components and mechanisms involved in this regulation is providing targets for the development of new antiviral therapies. Initiation of infection by herpes simplex virus (HSV) requires the activity of several cellular chromatin modification enzymes including the histone demethylases LSD1 and the family of JMJD2 proteins that promote transcriptional activation of the initial set of viral genes. Depletion of the JMJD2 members or inhibition of their activity with a new drug results in repression of expression of viral immediate early genes and abrogation of infection. This inhibitor also represses the reactivation of HSV from the latent state in sensory neurons. Like HSV, the ß-herpesvirus human cytomegalovirus also requires the activity of LSD1 and the JMJD2s to initiate infection, thus demonstrating the potential of this chromatin-based inhibitor to be useful against a variety of different viruses.


Assuntos
Epigênese Genética , Herpes Simples/prevenção & controle , Histona Desmetilases com o Domínio Jumonji/metabolismo , Simplexvirus/fisiologia , Ativação Viral , Latência Viral , Catálise , Regulação Viral da Expressão Gênica , Genes Precoces , Humanos , Regiões Promotoras Genéticas , Simplexvirus/genética
10.
Proc Natl Acad Sci U S A ; 108(7): 2747-52, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21285374

RESUMO

Host Cell Factor 1 (HCF-1) plays critical roles in regulating gene expression in a plethora of physiological processes. HCF-1 is first synthesized as a precursor, and subsequently specifically proteolytically cleaved within a large middle region termed the proteolytic processing domain (PPD). Although the underlying mechanism remains enigmatic, proteolysis of HCF-1 regulates its transcriptional activity and is important for cell cycle progression. Here we report that HCF-1 proteolysis is a regulated process. We demonstrate that a large proportion of the signaling enzyme O-linked-N-acetylglucosaminyl transferase (OGT) is complexed with HCF-1 and this interaction is essential for HCF-1 cleavage. Moreover, HCF-1 is, in turn, required for stabilizing OGT in the nucleus. We provide evidence indicating that OGT regulates HCF-1 cleavage via interaction with and O-GlcNAcylation of the HCF-1 PPD. In contrast, although OGT also interacts with the basic domain in the HCF-1 amino-terminal subunit, neither the interaction nor the O-GlcNAcylation of this region are required for proteolysis. Moreover, we show that OGT-mediated modulation of HCF-1 impacts the expression of the herpes simplex virus immediate-early genes, targets of HCF-1 during the initiation of viral infection. Together the data indicate that O-GlcNAcylation of HCF-1 is a signal for its proteolytic processing and reveal a unique crosstalk between these posttranslational modifications. Additionally, interactions of OGT with multiple HCF-1 domains may indicate that OGT has several functions in association with HCF-1.


Assuntos
Núcleo Celular/metabolismo , Regulação da Expressão Gênica/fisiologia , Fator C1 de Célula Hospedeira/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Western Blotting , Linhagem Celular , Imunoprecipitação da Cromatina , Imunofluorescência , Regulação da Expressão Gênica/genética , Humanos , Proteínas Imediatamente Precoces/metabolismo , Imunoprecipitação , Mutagênese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Simplexvirus/metabolismo
11.
J Biol Chem ; 285(18): 13364-71, 2010 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-20200153

RESUMO

THAP1 is a sequence-specific DNA binding factor that regulates cell proliferation through modulation of target genes such as the cell cycle-specific gene RRM1. Mutations in the THAP1 DNA binding domain, an atypical zinc finger (THAP-zf), have recently been found to cause DYT6 dystonia, a neurological disease characterized by twisting movements and abnormal postures. In this study, we report that THAP1 shares sequence characteristics, in vivo expression patterns and protein partners with THAP3, another THAP-zf protein. Proteomic analyses identified HCF-1, a potent transcriptional coactivator and cell cycle regulator, and O-GlcNAc transferase (OGT), the enzyme that catalyzes the addition of O-GlcNAc, as major cellular partners of THAP3. THAP3 interacts with HCF-1 through a consensus HCF-1-binding motif (HBM), a motif that is also present in THAP1. Accordingly, THAP1 was found to bind HCF-1 in vitro and to associate with HCF-1 and OGT in vivo. THAP1 and THAP3 belong to a large family of HCF-1 binding factors since seven other members of the human THAP-zf protein family were identified, which harbor evolutionary conserved HBMs and bind to HCF-1. Chromatin immunoprecipitation (ChIP) assays and RNA interference experiments showed that endogenous THAP1 mediates the recruitment of HCF-1 to the RRM1 promoter during endothelial cell proliferation and that HCF-1 is essential for transcriptional activation of RRM1. Together, our findings suggest HCF-1 is an important cofactor for THAP1. Interestingly, our results also provide an unexpected link between DYT6 and DYT3 (X-linked dystonia-parkinsonism) dystonias because the gene encoding the THAP1/DYT6 protein partner OGT maps within the DYT3 critical region on Xq13.1.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Cromossomos Humanos X/metabolismo , Proteínas de Ligação a DNA/metabolismo , Distonia/metabolismo , Fator C1 de Célula Hospedeira/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Proteínas Nucleares/metabolismo , Acetilglucosamina , Motivos de Aminoácidos , Proteínas Reguladoras de Apoptose/genética , Proliferação de Células , Cromossomos Humanos X/genética , Proteínas de Ligação a DNA/genética , Distonia/genética , Células Endoteliais , Doenças Genéticas Ligadas ao Cromossomo X , Células HeLa , Fator C1 de Célula Hospedeira/genética , Humanos , N-Acetilglucosaminiltransferases/genética , Proteínas Nucleares/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteômica , Ribonucleosídeo Difosfato Redutase , Transcrição Gênica/genética , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/genética , Dedos de Zinco
12.
Proc Natl Acad Sci U S A ; 107(6): 2461-6, 2010 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-20133788

RESUMO

The cellular transcriptional coactivator HCF-1 interacts with numerous transcription factors as well as other coactivators and is a component of multiple chromatin modulation complexes. The protein is essential for the expression of the immediate early genes of both herpes simplex virus (HSV) and varicella zoster virus and functions, in part, by coupling chromatin modification components including the Set1 or MLL1 histone methyltransferases and the histone demethylase LSD1 to promote the installation of positive chromatin marks and the activation of viral immediately early gene transcription. Although studies have investigated the role of HCF-1 in both cellular and viral transcription, little is known about other processes that the protein may be involved in. Here we demonstrate that HCF-1 localizes to sites of HSV replication late in infection. HCF-1 interacts directly and simultaneously with both HSV DNA replication proteins and the cellular histone chaperone Asf1b, a protein that regulates the progression of cellular DNA replication forks via chromatin reorganization. Asf1b localizes with HCF-1 in viral replication foci and depletion of Asf1b results in significantly reduced viral DNA accumulation. The results support a model in which the transcriptional coactivator HCF-1 is a component of the HSV DNA replication assembly and promotes viral DNA replication by coupling Asf1b to DNA replication components. This coupling provides a novel function for HCF-1 and insights into the mechanisms of modulating chromatin during DNA replication.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Replicação do DNA , Herpesvirus Humano 1/crescimento & desenvolvimento , Fator C1 de Célula Hospedeira/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Linhagem Celular , Chlorocebus aethiops , DNA Viral/genética , DNA Viral/metabolismo , Células HeLa , Herpesvirus Humano 1/genética , Histonas/metabolismo , Fator C1 de Célula Hospedeira/genética , Humanos , Imunoprecipitação , Microscopia de Fluorescência , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Ligação Proteica , Interferência de RNA , Técnicas do Sistema de Duplo-Híbrido , Células Vero , Proteínas Virais/genética , Proteínas Virais/metabolismo
13.
Biochim Biophys Acta ; 1799(3-4): 257-65, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19682612

RESUMO

The immediate early genes of the alpha-herpesviruses HSV and VZV are transcriptionally regulated by viral and cellular factors in a complex combinatorial manner. Despite this complexity and the apparent redundancy of activators, the expression of the viral IE genes is critically dependent upon the cellular transcriptional coactivator HCF-1. Although the role of HCF-1 had remained elusive, recent studies have demonstrated that the protein is a component of multiple chromatin modification complexes including the Set1/MLL1 histone H3K4 methyltransferases. Studies using model viral promoter-reporter systems as well as analyses of components recruited to the viral genome during the initiation of infection have elucidated the significance of HCF-1 chromatin modification complexes in contributing to the final state of modified histones assembled on the viral IE promoters. Strikingly, the absence of HCF-1 results in the accumulation of nucleosomes bearing repressive marks on the viral IE promoters and silencing of viral gene expression.


Assuntos
Alphaherpesvirinae/fisiologia , Cromatina/metabolismo , Regulação Viral da Expressão Gênica/fisiologia , Genes Precoces/genética , Fator C1 de Célula Hospedeira/metabolismo , Metilação de DNA , Histonas/metabolismo , Fator C1 de Célula Hospedeira/genética , Humanos , Transcrição Gênica
14.
Nat Med ; 15(11): 1312-7, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19855399

RESUMO

Reversible methylation of histone tails serves as either a positive signal recognized by transcriptional assemblies or a negative signal that result in repression. Invading viral pathogens that depend upon the host cell's transcriptional apparatus are also subject to the regulatory impact of chromatin assembly and modifications. Here we show that infection by the alpha-herpesviruses, herpes simplex virus (HSV) and varicella zoster virus (VZV), results in the rapid accumulation of chromatin bearing repressive histone H3 Lys9 methylation. To enable expression of viral immediate early (IE) genes, both viruses use the cellular transcriptional coactivator host cell factor-1 (HCF-1) to recruit the lysine-specific demethylase-1 (LSD1) to the viral immediate early promoters. Depletion of LSD1 or inhibition of its activity with monoamine oxidase inhibitors (MAOIs) results in the accumulation of repressive chromatin and a block to viral gene expression. As HCF-1 is a component of the Set1 and MLL1 histone H3 Lys4 methyltransferase complexes, it thus coordinates modulation of repressive H3 Lys9 methylation levels with addition of activating H3 Lys4 trimethylation marks. Strikingly, MAOIs also block the reactivation of HSV from latency in sensory neurons, indicating that the HCF-1 complex is a crucial component of the reactivation mechanism. The results support pharmaceutical control of histone modifying enzymes as a strategy for controlling herpesvirus infections.


Assuntos
Herpesvirus Humano 1/fisiologia , Histona Desmetilases/antagonistas & inibidores , Histona Desmetilases/metabolismo , Replicação Viral/fisiologia , Animais , Linhagem Celular Transformada , Imunoprecipitação da Cromatina/métodos , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/fisiologia , Ativação Enzimática/efeitos dos fármacos , Gânglios/citologia , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Regulação Viral da Expressão Gênica/fisiologia , Proteínas de Fluorescência Verde/genética , Herpesvirus Humano 3/fisiologia , Histona Desmetilases/genética , Histona Metiltransferases , Histona-Lisina N-Metiltransferase , Fator C1 de Célula Hospedeira/metabolismo , Humanos , Lisina/metabolismo , Camundongos , Inibidores da Monoaminoxidase/farmacologia , Mutação/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Pargilina/farmacologia , Fenciclidina/análogos & derivados , Piperidinas/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/fisiologia , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/farmacologia , Tempo de Reação/efeitos dos fármacos , Simplexvirus/fisiologia , Tiofenos/farmacologia , Fatores de Tempo , Transfecção/métodos , Replicação Viral/efeitos dos fármacos
15.
Proc Natl Acad Sci U S A ; 103(18): 6817-22, 2006 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-16624878

RESUMO

Limited proteolytic processing is an important transcriptional regulatory mechanism. In various contexts, proteolysis controls the cytoplasmic-to-nuclear transport of important transcription factors or removes domains to produce factors with altered activities. The transcriptional coactivator host cell factor-1 (HCF-1) is proteolytically processed within a unique domain consisting of 20-aa reiterations. Site-specific cleavage within one or more repeats generates a family of amino- and carboxyl-terminal subunits that remain tightly associated. However, the consequences of HCF-1 processing have been undefined. In this study, it was determined that the HCF-1-processing domain interacts with several proteins including the transcriptional coactivator/corepressor four-and-a-half LIM domain-2 (FHL2). Analysis of this interaction has uncovered specificity with both sequence and context determinants within the reiterations of this processing domain. In cells, FHL2 interacts exclusively with the nonprocessed coactivator and costimulates transcription of an HCF-1-dependent target gene. The functional interaction of HCF-1 with FHL2 supports a model in which site-specific proteolysis regulates the interaction of HCF-1 with protein partners and thus can modulate the activity of this coactivator. This paradigm expands the biological significance of limited proteolytic processing as a regulatory mechanism in gene transcription.


Assuntos
Proteínas de Homeodomínio/metabolismo , Fator C1 de Célula Hospedeira/metabolismo , Proteínas Musculares/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Sequência de Aminoácidos , Genes Reporter , Células HeLa , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Proteínas de Homeodomínio/genética , Fator C1 de Célula Hospedeira/genética , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Proteínas com Homeodomínio LIM , Dados de Sequência Molecular , Proteínas Musculares/genética , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição/genética , Técnicas do Sistema de Duplo-Híbrido , Proteínas Virais/genética , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...