Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
bioRxiv ; 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38746429

RESUMEN

Many stressors, including viral infection, induce a widespread suppression of cellular RNA polymerase II (RNAPII) transcription, yet the mechanisms underlying transcriptional repression are not well understood. Here we find that a crucial component of the RNA polymerase II holoenzyme, general transcription factor IIB (TFIIB), is targeted for post-translational turnover by two pathways, each of which contribute to its depletion during stress. Upon DNA damage, translational stress, apoptosis, or replication of the oncogenic Kaposi's sarcoma-associated herpesvirus (KSHV), TFIIB is cleaved by activated caspase-3, leading to preferential downregulation of pro-survival genes. TFIIB is further targeted for rapid proteasome-mediated turnover by the E3 ubiquitin ligase TRIM28. KSHV counteracts proteasome-mediated turnover of TFIIB, thereby preserving a sufficient pool of TFIIB for transcription of viral genes. Thus, TFIIB may be a lynchpin for transcriptional outcomes during stress and a key target for nuclear replicating DNA viruses that rely on host transcriptional machinery. Significance Statement: Transcription by RNA polymerase II (RNAPII) synthesizes all cellular protein-coding mRNA. Many cellular stressors and viral infections dampen RNAPII activity, though the processes underlying this are not fully understood. Here we describe a two-pronged degradation strategy by which cells respond to stress by depleting the abundance of the key RNAPII general transcription factor, TFIIB. We further demonstrate that an oncogenic human gammaherpesvirus antagonizes this process, retaining enough TFIIB to support its own robust viral transcription. Thus, modulation of RNAPII machinery plays a crucial role in dictating the outcome of cellular perturbation.

3.
bioRxiv ; 2023 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-37461644

RESUMEN

Gene regulation in eukaryotes relies on many mechanisms for optimal expression, including both protein transcription factors and DNA regulatory elements. CRISPR-based screens of both protein coding genes and non-coding regions have allowed identification of these transcriptional networks in human cells. Double-stranded DNA viruses also invoke human-like regulation to control transcription of viral genes that are required at different stages of the viral lifecycle. Here, we applied CRISPR-based tools to dissect regulation of a viral gene at high resolution in the oncogenic human herpesvirus Kaposi's sarcoma-associated herpesvirus (KSHV), whose compact, densely encoded genome provides unique challenges and opportunities for studying transcriptional networks. Through a combination of CRISPR-interference (CRISPRi) and Cas9 nuclease screening, we mapped a novel regulatory network comprised of coding and noncoding elements that influence expression of the essential KSHV protein ORF68 at early and late stages of the viral lifecycle. ORF68 encodes an essential protein involved in packaging the replicated viral DNA into nascent capsids. Although ORF68 expression initiates early in the viral lifecycle, we found that it is primarily required at later times. This work demonstrates the ability to exhaustively identify features controlling a given locus, capturing a complete viral regulatory circuit that functions within the human nucleus to control transcription.

4.
Cell Syst ; 14(6): 482-500.e8, 2023 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-37348463

RESUMEN

Viruses encode transcriptional regulatory proteins critical for controlling viral and host gene expression. Given their multifunctional nature and high sequence divergence, it is unclear which viral proteins can affect transcription and which specific sequences contribute to this function. Using a high-throughput assay, we measured the transcriptional regulatory potential of over 60,000 protein tiles across ∼1,500 proteins from 11 coronaviruses and all nine human herpesviruses. We discovered hundreds of transcriptional effector domains, including a conserved repression domain in all coronavirus Spike homologs, dual activation-repression domains in viral interferon regulatory factors (VIRFs), and an activation domain in six herpesvirus homologs of the single-stranded DNA-binding protein that we show is important for viral replication and late gene expression in Kaposi's sarcoma-associated herpesvirus (KSHV). For the effector domains we identified, we investigated their mechanisms via high-throughput sequence and chemical perturbations, pinpointing sequence motifs essential for function. This work massively expands viral protein annotations, serving as a springboard for studying their biological and health implications and providing new candidates for compact gene regulation tools.


Asunto(s)
Herpesvirus Humano 8 , Humanos , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Replicación Viral/genética , Regulación de la Expresión Génica
5.
PLoS Pathog ; 19(4): e1011163, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37068108

RESUMEN

ß- and γ-herpesviruses transcribe their late genes in a manner distinct from host transcription. This process is directed by a complex of viral transcriptional activator proteins that hijack cellular RNA polymerase II and an unknown set of additional factors. We employed proximity labeling coupled with mass spectrometry, followed by CRISPR and siRNA screening to identify proteins functionally associated with the Kaposi's sarcoma-associated herpesvirus (KSHV) late gene transcriptional complex. These data revealed that the catalytic subunit of the viral DNA packaging motor, ORF29, is both dynamically associated with the viral transcriptional activator complex and potentiates gene expression late in infection. Through genetic mutation and deletion of ORF29, we establish that its catalytic activity potentiates viral transcription and is required for robust accumulation of essential late proteins during infection. Thus, we propose an expanded role for ORF29 that encompasses its established function in viral packaging and its newly discovered contributions to viral transcription and late gene expression in KSHV.


Asunto(s)
Herpesvirus Humano 8 , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Empaquetamiento del Genoma Viral , Replicación Viral , Proteínas Virales/genética , Proteínas Virales/metabolismo , Expresión Génica , Regulación Viral de la Expresión Génica
6.
Microbiol Spectr ; : e0017223, 2023 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-36752632

RESUMEN

RNA polymerase III (RNAPIII) transcribes a variety of noncoding RNAs, including tRNA (tRNA) and the B2 family of short interspersed nuclear elements (SINEs). B2 SINEs are noncoding retrotransposons that possess tRNA-like promoters and are normally silenced in healthy somatic tissue. Infection with the murine gammaherpesvirus MHV68 induces transcription of both SINEs and tRNAs, in part through the activity of the viral protein kinase ORF36. Here, we identify the conserved MHV68 tegument protein ORF45 as an additional activator of these RNAPIII loci. MHV68 ORF45 and ORF36 form a complex, resulting in an additive induction RNAPIII and increased ORF45 expression. ORF45-induced RNAPIII transcription is dependent on its activation of the extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK) signaling pathway, which in turn increases the abundance of the RNAPIII transcription factor Brf1. Other viral and nonviral activators of MAPK/ERK signaling also increase the levels of Brf1 protein, B2 SINE RNA, and tRNA, suggesting that this is a common strategy to increase RNAPIII activity. IMPORTANCE Gammaherpesviral infection alters the gene expression landscape of a host cell, including through the induction of noncoding RNAs transcribed by RNA polymerase III (RNAPIII). Among these are a class of repetitive genes known as retrotransposons, which are normally silenced elements and can copy and spread throughout the genome, and transfer RNAs (tRNAs), which are fundamental components of protein translation machinery. How these loci are activated during infection is not well understood. Here, we identify ORF45 from the model murine gammaherpesvirus MHV68 as a novel activator of RNAPIII transcription. To do so, it engages the MAPK/ERK signaling pathway, which is a central regulator of cellular response to environmental stimuli. Activation of this pathway leads to the upregulation of a key factor required for RNAPIII activity, Brf1. These findings expand our understanding of the regulation and dysregulation of RNAPIII transcription and highlight how viral cooption of key signaling pathways can impact host gene expression.

7.
Nucleic Acids Res ; 51(1): 182-197, 2023 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-36537232

RESUMEN

Alkaline exonucleases (AE) are present in several large DNA viruses including bacteriophage λ and herpesviruses, where they play roles in viral DNA processing during genome replication. Given the genetic conservation of AEs across viruses infecting different kingdoms of life, these enzymes likely assume central roles in the lifecycles of viruses where they have yet to be well characterized. Here, we applied a structure-guided functional analysis of the bifunctional AE in the oncogenic human gammaherpesvirus Kaposi's sarcoma-associated herpesvirus (KSHV), called SOX. In addition to identifying a preferred DNA substrate preference for SOX, we define key residues important for DNA binding and DNA processing, and how SOX activity on DNA partially overlaps with its functionally separable cleavage of mRNA. By engineering these SOX mutants into KSHV, we reveal roles for its DNase activity in viral gene expression and infectious virion production. Our results provide mechanistic insight into gammaherpesviral AE activity as well as areas of functional conservation between this mammalian virus AE and its distant relative in phage λ.


Asunto(s)
Exonucleasas , Herpesvirus Humano 8 , Animales , Humanos , ADN Viral/metabolismo , Exonucleasas/genética , Expresión Génica , Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/metabolismo , Mamíferos/genética , Virión/metabolismo , Replicación Viral
8.
Curr Opin Virol ; 56: 101259, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36162260

RESUMEN

Control of gene expression, including transcription, is central in dictating the outcome of viral infection. One of the profound alterations induced by viruses is modification to the integrity and function of eukaryotic RNA polymerase II (Pol II). Here, we discuss how infection perturbs the Pol II complex by altering subunit phosphorylation and turnover, as well as how cellular genotoxic stress (e.g. DNA damage) elicits similar outcomes. By highlighting emerging parallels and differences in Pol II control during viral infection and abiotic stress, we hope to bolster identification of pathways that target Pol II and regulate the transcriptome.

9.
PLoS Pathog ; 18(2): e1010099, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35202449

RESUMEN

The mRNA 5' cap structure serves both to protect transcripts from degradation and promote their translation. Cap removal is thus an integral component of mRNA turnover that is carried out by cellular decapping enzymes, whose activity is tightly regulated and coupled to other stages of the mRNA decay pathway. The poxvirus vaccinia virus (VACV) encodes its own decapping enzymes, D9 and D10, that act on cellular and viral mRNA, but may be regulated differently than their cellular counterparts. Here, we evaluated the targeting potential of these viral enzymes using RNA sequencing from cells infected with wild-type and decapping mutant versions of VACV as well as in uninfected cells expressing D10. We found that D9 and D10 target an overlapping subset of viral transcripts but that D10 plays a dominant role in depleting the vast majority of human transcripts, although not in an indiscriminate manner. Unexpectedly, the splicing architecture of a gene influences how robustly its corresponding transcript is targeted by D10, as transcripts derived from intronless genes are less susceptible to enzymatic decapping by D10. As all VACV genes are intronless, preferential decapping of transcripts from intron-containing genes provides an unanticipated mechanism for the virus to disproportionately deplete host transcripts and remodel the infected cell transcriptome.


Asunto(s)
Poxviridae , Virus Vaccinia , Endorribonucleasas/metabolismo , Humanos , Poxviridae/genética , Caperuzas de ARN/genética , Caperuzas de ARN/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Virus Vaccinia/genética , Virus Vaccinia/metabolismo , Proteínas Virales/metabolismo
10.
Nat Commun ; 13(1): 623, 2022 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-35110532

RESUMEN

RNA polymerase III (Pol III) transcribes noncoding RNA, including transfer RNA (tRNA), and is commonly targeted during cancer and viral infection. We find that Herpes Simplex Virus-1 (HSV-1) stimulates tRNA expression 10-fold. Perturbation of host tRNA synthesis requires nuclear viral entry, but not synthesis of specific viral transcripts. tRNA with a specific codon bias were not targeted-rather increased transcription was observed from euchromatic, actively transcribed loci. tRNA upregulation is linked to unique crosstalk between the Pol II and III transcriptional machinery. While viral infection results in depletion of Pol II on host mRNA promoters, we find that Pol II binding to tRNA loci increases. Finally, we report Pol III and associated factors bind the viral genome, which suggests a previously unrecognized role in HSV-1 gene expression. These findings provide insight into mechanisms by which HSV-1 alters the host nuclear environment, shifting key processes in favor of the pathogen.


Asunto(s)
Herpesvirus Humano 1/fisiología , ARN Polimerasa III/metabolismo , Factores de Transcripción , Genoma Viral , Herpesvirus Humano 1/genética , Humanos , Regiones Promotoras Genéticas , ARN Polimerasa II/metabolismo , ARN Polimerasa III/genética , ARN Mensajero/metabolismo , ARN de Transferencia , ARN no Traducido , Transcripción Genética , Activación Transcripcional , Replicación Viral
11.
PLoS Pathog ; 18(1): e1010236, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35041709

RESUMEN

While traditional methods for studying large DNA viruses allow the creation of individual mutants, CRISPR/Cas9 can be used to rapidly create thousands of mutant dsDNA viruses in parallel, enabling the pooled screening of entire viral genomes. Here, we applied this approach to Kaposi's sarcoma-associated herpesvirus (KSHV) by designing a sgRNA library containing all possible ~22,000 guides targeting the 154 kilobase viral genome, corresponding to one cut site approximately every 8 base pairs. We used the library to profile viral sequences involved in transcriptional activation of late genes, whose regulation involves several well characterized features including dependence on viral DNA replication and a known set of viral transcriptional activators. Upon phenotyping all possible Cas9-targeted viruses for transcription of KSHV late genes we recovered these established regulators and identified a new required factor (ORF46), highlighting the utility of the screening pipeline. By performing targeted deep sequencing of the viral genome to distinguish between knock-out and in-frame alleles created by Cas9, we identify the DNA binding but not catalytic domain of ORF46 to be required for viral DNA replication and thus late gene expression. Our pooled Cas9 tiling screen followed by targeted deep viral sequencing represents a two-tiered screening paradigm that may be widely applicable to dsDNA viruses.


Asunto(s)
Regulación Viral de la Expresión Génica/fisiología , Genes Virales/genética , Herpesvirus Humano 8/genética , Sistemas CRISPR-Cas , Células HEK293 , Humanos
13.
Cell Rep ; 37(3): 109841, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34624207

RESUMEN

Nonstructural protein 1 (nsp1) is a coronavirus (CoV) virulence factor that restricts cellular gene expression by inhibiting translation through blocking the mRNA entry channel of the 40S ribosomal subunit and by promoting mRNA degradation. We perform a detailed structure-guided mutational analysis of severe acute respiratory syndrome (SARS)-CoV-2 nsp1, revealing insights into how it coordinates these activities against host but not viral mRNA. We find that residues in the N-terminal and central regions of nsp1 not involved in docking into the 40S mRNA entry channel nonetheless stabilize its association with the ribosome and mRNA, both enhancing its restriction of host gene expression and enabling mRNA containing the SARS-CoV-2 leader sequence to escape translational repression. These data support a model in which viral mRNA binding functionally alters the association of nsp1 with the ribosome, which has implications for drug targeting and understanding how engineered or emerging mutations in SARS-CoV-2 nsp1 could attenuate the virus.


Asunto(s)
COVID-19/genética , Regulación Viral de la Expresión Génica , SARS-CoV-2/genética , Proteínas no Estructurales Virales/metabolismo , Anisotropía , COVID-19/inmunología , Análisis Mutacional de ADN , Femenino , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Cinética , Mutación , Fenotipo , Mutación Puntual , Biosíntesis de Proteínas , Dominios Proteicos , Estabilidad del ARN , Subunidades Ribosómicas Pequeñas de Eucariotas/metabolismo , Ribosomas/metabolismo
14.
Elife ; 102021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-34085923

RESUMEN

RNA abundance is generally sensitive to perturbations in decay and synthesis rates, but crosstalk between RNA polymerase II transcription and cytoplasmic mRNA degradation often leads to compensatory changes in gene expression. Here, we reveal that widespread mRNA decay during early apoptosis represses RNAPII transcription, indicative of positive (rather than compensatory) feedback. This repression requires active cytoplasmic mRNA degradation, which leads to impaired recruitment of components of the transcription preinitiation complex to promoter DNA. Importin α/ß-mediated nuclear import is critical for this feedback signaling, suggesting that proteins translocating between the cytoplasm and nucleus connect mRNA decay to transcription. We also show that an analogous pathway activated by viral nucleases similarly depends on nuclear protein import. Collectively, these data demonstrate that accelerated mRNA decay leads to the repression of mRNA transcription, thereby amplifying the shutdown of gene expression. This highlights a conserved gene regulatory mechanism by which cells respond to threats.


Asunto(s)
Apoptosis , Neoplasias/metabolismo , ARN Polimerasa II/metabolismo , Estabilidad del ARN , ARN Mensajero/metabolismo , ARN Neoplásico/metabolismo , Transcripción Genética , Transporte Activo de Núcleo Celular , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Citoplasma/genética , Citoplasma/metabolismo , Retroalimentación Fisiológica , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , ARN Polimerasa II/genética , ARN Mensajero/genética , ARN Neoplásico/genética , Factores de Tiempo , alfa Carioferinas/metabolismo , beta Carioferinas/metabolismo
15.
Elife ; 102021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33554858

RESUMEN

Genome packaging in large double-stranded DNA viruses requires a powerful molecular motor to force the viral genome into nascent capsids, which involves essential accessory factors that are poorly understood. Here, we present structures of two such accessory factors from the oncogenic herpesviruses Kaposi's sarcoma-associated herpesvirus (KSHV; ORF68) and Epstein-Barr virus (EBV; BFLF1). These homologous proteins form highly similar homopentameric rings with a positively charged central channel that binds double-stranded DNA. Mutation of individual positively charged residues within but not outside the channel ablates DNA binding, and in the context of KSHV infection, these mutants fail to package the viral genome or produce progeny virions. Thus, we propose a model in which ORF68 facilitates the transfer of newly replicated viral genomes to the packaging motor.


Asunto(s)
Herpesvirus Humano 4/fisiología , Herpesvirus Humano 8/fisiología , Proteínas Virales/química , Proteínas Virales/metabolismo , Infecciones por Virus de Epstein-Barr/virología , Genoma Viral , Células HEK293 , Infecciones por Herpesviridae/virología , Herpesvirus Humano 4/química , Herpesvirus Humano 4/genética , Herpesvirus Humano 8/química , Herpesvirus Humano 8/genética , Humanos , Empaquetamiento del Genoma Viral , Proteínas Virales/genética , Replicación Viral
16.
mBio ; 11(6)2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33323507

RESUMEN

Transfer RNAs (tRNAs) are transcribed by RNA polymerase III (RNAPIII) and play a central role in decoding our genome, yet their expression and noncanonical function remain understudied. Many DNA tumor viruses enhance the activity of RNAPIII, yet whether infection alters tRNA expression is largely unknown. Here, we present the first genome-wide analysis of how viral infection alters the tRNAome. Using a tRNA-specific sequencing method (DM-tRNA-seq), we find that the murine gammaherpesvirus MHV68 induces global changes in premature tRNA (pre-tRNA) expression, with 14% of tRNA genes upregulated more than 3-fold, indicating that differential tRNA gene induction is a characteristic of DNA virus infection. Elevated pre-tRNA expression corresponds to increased RNAPIII occupancy for the subset of tRNA genes tested; additionally, posttranscriptional mechanisms contribute to the accumulation of pre-tRNA species. We find increased abundance of tRNA fragments derived from pre-tRNAs upregulated by viral infection, suggesting that noncanonical tRNA cleavage is also affected. Furthermore, pre-tRNA accumulation, but not RNAPIII recruitment, requires gammaherpesvirus-induced degradation of host mRNAs by the virally encoded mRNA endonuclease muSOX. We hypothesize that depletion of pre-tRNA maturation or turnover machinery contributes to robust accumulation of full-length pre-tRNAs in infected cells. Collectively, these findings reveal pervasive changes to tRNA expression during DNA virus infection and highlight the potential of using viruses to explore tRNA biology.IMPORTANCE Viral infection can dramatically change the gene expression landscape of the host cell, yet little is known regarding changes in noncoding gene transcription by RNA polymerase III (RNAPIII). Among these are transfer RNAs (tRNAs), which are fundamental in protein translation, yet whose gene regulatory features remain largely undefined in mammalian cells. Here, we perform the first genome-wide analysis of tRNA expression changes during viral infection. We show that premature tRNAs accumulate during infection with the model gammaherpesvirus MHV68 as a consequence of increased transcription, but that transcripts do not undergo canonical maturation into mature tRNAs. These findings underscore how tRNA expression is a highly regulated process, especially during conditions of elevated RNAPIII activity.


Asunto(s)
Gammaherpesvirinae/fisiología , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/metabolismo , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Animales , Línea Celular , Gammaherpesvirinae/genética , Infecciones por Herpesviridae/virología , Humanos , Ratones , ARN Polimerasa III/genética , ARN Polimerasa III/metabolismo , Procesamiento Postranscripcional del ARN , Estabilidad del ARN , Transcripción Genética
17.
PLoS Pathog ; 16(9): e1008843, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32886723

RESUMEN

ß- and γ-herpesviruses include the oncogenic human viruses Kaposi's sarcoma-associated virus (KSHV) and Epstein-Barr virus (EBV), and human cytomegalovirus (HCMV), which is a significant cause of congenital disease. Near the end of their replication cycle, these viruses transcribe their late genes in a manner distinct from host transcription. Late gene transcription requires six virally encoded proteins, one of which is a functional mimic of host TATA-box-binding protein (TBP) that is also involved in recruitment of RNA polymerase II (Pol II) via unknown mechanisms. Here, we applied biochemical protein interaction studies together with electron microscopy-based imaging of a reconstituted human preinitiation complex to define the mechanism underlying Pol II recruitment. These data revealed that the herpesviral TBP, encoded by ORF24 in KSHV, makes a direct protein-protein contact with the C-terminal domain of host RNA polymerase II (Pol II), which is a unique feature that functionally distinguishes viral from cellular TBP. The interaction is mediated by the N-terminal domain (NTD) of ORF24 through a conserved motif that is shared in its ß- and γ-herpesvirus homologs. Thus, these herpesviruses employ an unprecedented strategy in eukaryotic transcription, wherein promoter recognition and polymerase recruitment are facilitated by a single transcriptional activator with functionally distinct domains.


Asunto(s)
Herpesvirus Humano 8/metabolismo , ARN Polimerasa II/metabolismo , Proteína de Unión a TATA-Box/metabolismo , Proteínas Virales/metabolismo , Secuencias de Aminoácidos , Células HEK293 , Herpesvirus Humano 8/genética , Humanos , Unión Proteica , Dominios Proteicos , ARN Polimerasa II/genética , Proteína de Unión a TATA-Box/genética , Proteínas Virales/genética
18.
J Biol Chem ; 295(37): 12910-12934, 2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32661197

RESUMEN

Few human pathogens have been the focus of as much concentrated worldwide attention as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the cause of COVID-19. Its emergence into the human population and ensuing pandemic came on the heels of severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV), two other highly pathogenic coronavirus spillovers, which collectively have reshaped our view of a virus family previously associated primarily with the common cold. It has placed intense pressure on the collective scientific community to develop therapeutics and vaccines, whose engineering relies on a detailed understanding of coronavirus biology. Here, we present the molecular virology of coronavirus infection, including its entry into cells, its remarkably sophisticated gene expression and replication mechanisms, its extensive remodeling of the intracellular environment, and its multifaceted immune evasion strategies. We highlight aspects of the viral life cycle that may be amenable to antiviral targeting as well as key features of its biology that await discovery.


Asunto(s)
Infecciones por Coronavirus/virología , Coronavirus/fisiología , Regulación Viral de la Expresión Génica , Fenómenos Fisiológicos de los Virus , Animales , Antígenos Virales/inmunología , Coronavirus/genética , Coronavirus/inmunología , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/inmunología , Interacciones Huésped-Patógeno , Humanos
19.
J Virol ; 94(14)2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32404524

RESUMEN

Short interspersed nuclear elements (SINEs) are RNA polymerase III (RNAPIII)-transcribed, retrotransposable noncoding RNA (ncRNA) elements ubiquitously spread throughout mammalian genomes. While normally silenced in healthy somatic tissue, SINEs can be induced during infection with DNA viruses, including the model murine gammaherpesvirus 68 (MHV68). Here, we explored the mechanisms underlying MHV68 activation of SINE ncRNAs. We demonstrate that lytic MHV68 infection of B cells, macrophages, and fibroblasts leads to robust activation of the B2 family of SINEs in a cell-autonomous manner. B2 ncRNA induction requires neither host innate immune signaling factors nor involvement of the RNAPIII master regulator Maf1. However, we identified MHV68 ORF36, the conserved herpesviral kinase, as playing a key role in B2 induction during lytic infection. SINE activation is linked to ORF36 kinase activity and can also be induced by inhibition of histone deacetylases 1 and 2 (HCAC 1/2), which is one of the known ORF36 functions. Collectively, our data suggest that ORF36-mediated changes in chromatin modification contribute to B2 activation during MHV68 infection and that this activity is conserved in other herpesviral protein kinase homologs.IMPORTANCE Viral infection dramatically changes the levels of many types of RNA in a cell. In particular, certain oncogenic viruses activate expression of repetitive genes called retrotransposons, which are normally silenced due to their ability to copy and spread throughout the genome. Here, we established that infection with the gammaherpesvirus MHV68 leads to a dramatic induction of a class of noncoding retrotransposons called B2 SINEs in multiple cell types. We then explored how MHV68 activates B2 SINEs, revealing a role for the conserved herpesviral protein kinase ORF36. Both ORF36 kinase-dependent and kinase-independent functions contribute to B2 induction, perhaps through ORF36 targeting of proteins involved in controlling the accessibility of chromatin surrounding SINE loci. Understanding the features underlying induction of these elements following MHV68 infection should provide insight into core elements of SINE regulation, as well as disregulation of SINE elements associated with disease.


Asunto(s)
Infecciones por Herpesviridae/enzimología , Proteínas Quinasas/metabolismo , Retroelementos , Rhadinovirus/enzimología , Proteínas Virales/metabolismo , Animales , Linfocitos B/enzimología , Linfocitos B/patología , Linfocitos B/virología , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/patología , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo , Macrófagos/enzimología , Macrófagos/patología , Macrófagos/virología , Ratones , Células 3T3 NIH , Proteínas Quinasas/genética , ARN no Traducido/genética , ARN no Traducido/metabolismo , Rhadinovirus/genética , Proteínas Virales/genética
20.
PLoS Pathog ; 16(2): e1008269, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32032393

RESUMEN

In mammalian cells, widespread acceleration of cytoplasmic mRNA degradation is linked to impaired RNA polymerase II (Pol II) transcription. This mRNA decay-induced transcriptional repression occurs during infection with gammaherpesviruses including Kaposi's sarcoma-associated herpesvirus (KSHV) and murine gammaherpesvirus 68 (MHV68), which encode an mRNA endonuclease that initiates widespread RNA decay. Here, we show that MHV68-induced mRNA decay leads to a genome-wide reduction of Pol II occupancy at mammalian promoters. This reduced Pol II occupancy is accompanied by down-regulation of multiple Pol II subunits and TFIIB in the nucleus of infected cells, as revealed by mass spectrometry-based global measurements of protein abundance. Viral genes, despite the fact that they require Pol II for transcription, escape transcriptional repression. Protection is not governed by viral promoter sequences; instead, location on the viral genome is both necessary and sufficient to escape the transcriptional repression effects of mRNA decay. We propose a model in which the ability to escape from transcriptional repression is linked to the localization of viral DNA within replication compartments, providing a means for these viruses to counteract decay-induced transcript loss.


Asunto(s)
Infecciones por Herpesviridae/metabolismo , Herpesvirus Humano 8/fisiología , Regiones Promotoras Genéticas , ARN Polimerasa II/metabolismo , Estabilidad del ARN , Rhadinovirus/fisiología , Replicación Viral , Animales , Endonucleasas/genética , Endonucleasas/metabolismo , Genoma Viral , Infecciones por Herpesviridae/genética , Ratones , Células 3T3 NIH , ARN Polimerasa II/genética , Factor de Transcripción TFIIB/genética , Factor de Transcripción TFIIB/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...