Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros











Intervalo de año de publicación
1.
Viruses ; 16(6)2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38932139

RESUMEN

The viral interferon regulatory factors (vIRFs) of KSHV are known to dysregulate cell signaling pathways to promote viral oncogenesis and to block antiviral immune responses to facilitate infection. However, it remains unknown to what extent each vIRF plays a role in gene regulation. To address this, we performed a comparative analysis of the protein structures and gene regulation of the four vIRFs. Our structure prediction analysis revealed that despite their low amino acid sequence similarity, vIRFs exhibit high structural homology in both their DNA-binding domain (DBD) and IRF association domain. However, despite this shared structural homology, we demonstrate that each vIRF regulates a distinct set of KSHV gene promoters and human genes in epithelial cells. We also found that the DBD of vIRF1 is essential in regulating the expression of its target genes. We propose that the structurally similar vIRFs evolved to possess specialized transcriptional functions to regulate specific genes.


Asunto(s)
Células Epiteliales , Regulación Viral de la Expresión Génica , Herpesvirus Humano 8 , Factores Reguladores del Interferón , Proteínas Virales , Humanos , Factores Reguladores del Interferón/metabolismo , Factores Reguladores del Interferón/genética , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/fisiología , Células Epiteliales/virología , Proteínas Virales/metabolismo , Proteínas Virales/genética , Regiones Promotoras Genéticas , Transcripción Genética , Genoma Viral , Línea Celular
2.
J Virol ; 98(6): e0025524, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38752725

RESUMEN

Human herpesvirus 8 (HHV-8), associated with Kaposi sarcoma, primary effusion lymphoma (PEL), and multicentric Castleman disease, encodes four interferon regulatory factor homologs, vIRFs 1-4, that interact with and inhibit various mediators of host-cell defense against virus infection. A cellular protein targeted by all the vIRFs is ubiquitin-specific protease 7 (USP7); while replication-modulatory and latently infected PEL-cell pro-viability phenotypes of USP7 targeting have been identified for vIRFs 1-3, the significance of the interaction of vIRF-4 with USP7 has remained undetermined. Here we show, through genetic ablation of the vIRF-4-USP7 interaction in infected cells, that vIRF-4 association with USP7 is necessary for optimal expression of vIRF-4 and normal HHV-8 replication. Findings from experiments on transfected and infected cells identified ubiquitination of vIRF-4 via K48-linkage and USP7-binding-associated suppression of vIRF-4 ubiquitination and, in infected cells, increased vIRF-4 expression. Analysis of IFN-I induction and associated signaling as a function of vIRF-4 and its interaction with USP7 identified a role of each in innate-immune suppression. Finally, activation via K63-polyubiquitination of the innate-immune signaling mediator TRAF3 was found to be suppressed by vIRF-4 in a USP7-binding-associated manner in infected cells, but not in transfected cells, likely via binding-regulated expression of vIRF-4. Together, our data identify the first examples of vIRF ubiquitination and a vIRF substrate of USP7, enhanced expression of vIRF-4 via its interaction with USP7, and TRAF3-inhibitory activity of vIRF-4. The findings address, for the first time, the biological significance of the interaction of vIRF-4 with USP7 and reveal a mechanism of vIRF-4-mediated innate-immune evasion and pro-replication activity via TRAF3 regulation. IMPORTANCE: HHV-8 homologs of cellular interferon regulatory factors (IRFs), involved in host-cell defense against virus infection, interact in an inhibitory fashion with IRFs and other mediators of antiviral innate immunity. These interactions are of demonstrated or hypothesized importance for successful primary, productive (lytic), and latent (persistent) infection by HHV-8. While HHV-8 vIRF-4 is known to interact physically with USP7 deubiquitinase, a key regulator of various cellular proteins, the functional and biological significance of the interaction has not been addressed. The present study identifies the interaction as important for HHV-8 productive replication and, indeed, for vIRF-4 expression and reveals a new function of vIRF-4 via inhibition of the activity of TRAF3, a pivotal mediator of host-cell antiviral activity through activation of cellular IRFs and induction of type-I interferons. These findings identify potential targets for the development of novel anti-HHV-8 agents, such as those able to disrupt vIRF-4-USP7 interaction or vIRF-4-stabilizing USP7 activity.


Asunto(s)
Herpesvirus Humano 8 , Factores Reguladores del Interferón , Peptidasa Específica de Ubiquitina 7 , Ubiquitinación , Proteínas Virales , Replicación Viral , Humanos , Herpesvirus Humano 8/fisiología , Herpesvirus Humano 8/metabolismo , Peptidasa Específica de Ubiquitina 7/metabolismo , Peptidasa Específica de Ubiquitina 7/genética , Factores Reguladores del Interferón/metabolismo , Factores Reguladores del Interferón/genética , Proteínas Virales/metabolismo , Proteínas Virales/genética , Células HEK293 , Factor 3 Asociado a Receptor de TNF/metabolismo , Factor 3 Asociado a Receptor de TNF/genética , Unión Proteica , Interacciones Huésped-Patógeno
3.
Microorganisms ; 11(7)2023 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-37512882

RESUMEN

Shigella spp. cause hundreds of millions of intestinal infections each year. They target the mucosa of the human colon and are an important model of intracellular bacterial pathogenesis. Shigella is a pathovar of Escherichia coli that is characterized by the presence of a large invasion plasmid, pINV, which encodes the characteristic type III secretion system and icsA used for cytosol invasion and cell-to-cell spread, respectively. First, we review recent advances in the genetic aspects of Shigella, shedding light on its evolutionary history within the E. coli lineage and its relationship to the acquisition of pINV. We then discuss recent insights into the processes that allow for the maintenance of pINV. Finally, we describe the role of the transcription activators VirF, VirB, and MxiE in the major virulence gene regulatory cascades that control the expression of the type III secretion system and icsA. This provides an opportunity to examine the interplay between these pINV-encoded transcriptional activators and numerous chromosome-encoded factors that modulate their activity. Finally, we discuss novel chromosomal genes icaR, icaT, and yccE that are regulated by MxiE. This review emphasizes the notion that Shigella and E. coli have walked the fine line between commensalism and pathogenesis for much of their history.

4.
Autophagy Rep ; 2(1)2023.
Artículo en Inglés | MEDLINE | ID: mdl-38214011

RESUMEN

Viral control of mitochondria via mitophagy has a dampening effect on mitochondrion-mediated innate immune responses. We previously found that human herpesvirus 8 (HHV-8) could activate mitophagy via its lytic gene product vIRF-1 (viral interferon regulatory factor 1). Mechanistically, we previously demonstrated that vIRF-1 interacts with the mitophagic proteins BNIP3L (BCL2 interacting protein 3 like) and TUFM (Tu translation elongation factor, mitochondrial). Despite these significant findings, however, the precise molecular mechanisms underlying vIRF-1-activated mitophagy, particularly with core components of the autophagy machinery, remained to be fully elucidated. We recently reported that vIRF-1 binds preferentially and directly to GABARAPL1 (GABA type A receptor associated protein like 1) in a noncanonical manner, and this interaction is essential for virus-productive replication. Furthermore, we found that BNIP3L is a crucial factor that promotes vIRF-1 oligomerization and associated mitophagy activation, including GABARAPL1 interaction with vIRF-1 and TUFM dimerization. Together, our findings deepen our understanding of lytic infection-induced mitophagy and provide the key protein-protein interactions involved in vIRF-1-mediated mitophagy.

5.
J Bacteriol ; 204(7): e0013722, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35703565

RESUMEN

Members of the AraC family of transcriptional regulators (AFTRs) control the expression of many genes important to cellular processes, including virulence. In Shigella species, the type III secretion system (T3SS), a key determinant for host cell invasion, is regulated by the three-tiered VirF/VirB/MxiE transcriptional cascade. Both VirF and MxiE belong to the AFTRs and are characterized as positive transcriptional regulators. Here, we identify a novel regulatory activity for MxiE and its coregulator IpgC, which manifests as a negative feedback loop in the VirF/VirB/MxiE transcriptional cascade. Our findings show that MxiE and IpgC downregulate the virB promoter and, hence, VirB protein production, thus decreasing VirB-dependent promoter activity at ospD1, one of the nearly 50 VirB-dependent genes. At the virB promoter, regions required for negative MxiE- and IpgC-dependent regulation were mapped and found to be coincident with regions required for positive VirF-dependent regulation. In tandem, negative MxiE- and IpgC-dependent regulation of the virB promoter only occurred in the presence of VirF, suggesting that MxiE and IpgC can function to counter VirF activation of the virB promoter. Lastly, MxiE and IpgC do not downregulate another VirF-activated promoter, icsA, demonstrating that this negative feedback loop targets the virB promoter. Our study provides insight into a mechanism that may reprogram Shigella virulence gene expression following type III secretion and provides the impetus to examine if MxiE and IpgC homologs in other important bacterial pathogens, such as Burkholderia pseudomallei and Salmonella enterica serovars Typhimurium and Typhi, coordinate similar negative feedback loops. IMPORTANCE The large AraC family of transcriptional regulators (AFTRs) control virulence gene expression in many bacterial pathogens. In Shigella species, the AraC/XylS protein MxiE and its coregulator IpgC positively regulate the expression of type III secretion system genes within the three-tiered VirF/VirB/MxiE transcriptional cascade. Our findings suggest a negative feedback loop in the VirF/VirB/MxiE cascade, in which MxiE and IpgC counter VirF-dependent activation of the virB promoter, thus making this the first characterization of negative MxiE- and IpgC-dependent regulation. Our study provides insight into a mechanism that likely reprograms Shigella virulence gene expression following type III secretion, which has implications for other important bacterial pathogens with functional homologs of MxiE and IpgC.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Shigella flexneri , Proteínas Bacterianas/metabolismo , Citarabina/metabolismo , Proteínas de Unión al ADN/metabolismo , Retroalimentación , Shigella flexneri/genética , Shigella flexneri/metabolismo , Transcripción Genética , Sistemas de Secreción Tipo III/genética , Sistemas de Secreción Tipo III/metabolismo , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
6.
Virulence ; 12(1): 1661-1671, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34152261

RESUMEN

Shigella is an intracellular pathogen that primarily infects the human colon and causes shigellosis. Shigella virulence relies largely on the type III secretion system (T3SS) and secreted effectors. VirF, the master Shigella virulence regulator, is essential for the expression of T3SS-related genes. In this study, we found that YhjC, a LysR-type transcriptional regulator, is required for Shigella virulence through activating the transcription of virF. Pathogenicity of the yhjC mutant, including colonization in the colons of guinea pigs as well as its ability for host cell adhesion and invasion, was significantly lowered. Expression levels of virF and nearly all VirF-dependent genes were downregulated by yhjC deletion, indicating that YhjC can activate virF transcription. Electrophoretic mobility shift assay analysis demonstrated that YhjC could bind directly to the virF promoter region. Therefore, YhjC is a novel virulence regulator that positively regulates the virF expression and promotes Shigella virulence. Additionally, genome-wide expression analysis identified the presence of other genes in the large virulence plasmid and a genome exhibiting differential expression in response to yhjC deletion, with 169 downregulated and 99 upregulated genes, indicating that YhjC also functioned as a global regulatory factor.


Asunto(s)
Proteínas Bacterianas , Regulación Bacteriana de la Expresión Génica , Shigella flexneri , Factores de Virulencia , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Cobayas , Plásmidos , Shigella flexneri/genética , Shigella flexneri/metabolismo , Transcripción Genética , Sistemas de Secreción Tipo III , Virulencia , Factores de Virulencia/genética
7.
J Virol ; 95(9)2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33597212

RESUMEN

Studies on "hit-and-run" effects by viral proteins are difficult when using traditional affinity precipitation-based techniques under dynamic conditions, because only proteins interacting at a specific instance in time can be precipitated by affinity purification. Recent advances in proximity labeling (PL) have enabled identification of both static and dynamic protein-protein interactions. In this study, we applied a PL method by generating recombinant Kaposi's sarcoma-associated herpesvirus (KSHV). KSHV, a gammaherpesvirus, uniquely encodes four interferon regulatory factors (IRF-1 to -4) that suppress host interferon responses, and we examined KSHV IRF-1 and IRF-4 neighbor proteins to identify cellular proteins involved in innate immune regulation. PL identified 213 and 70 proteins as neighboring proteins of viral IRF-1 (vIRF-1) and vIRF-4 during viral reactivation, and 47 proteins were shared between the two vIRFs; the list also includes three viral proteins, ORF17, thymidine kinase, and vIRF-4. Functional annotation of respective interacting proteins showed highly overlapping biological roles such as mRNA processing and transcriptional regulation by TP53. Innate immune regulation by these commonly interacting 44 cellular proteins was examined with small interfering RNAs (siRNAs), and the splicing factor 3B family proteins were found to be associated with interferon transcription and to act as suppressors of KSHV reactivation. We propose that recombinant mini-TurboID-KSHV is a powerful tool to probe key cellular proteins that play a role in KSHV replication and that selective splicing factors have a function in the regulation of innate immune responses.IMPORTANCE Viral protein interaction with a host protein shows at least two sides: (i) taking host protein functions for its own benefit and (ii) disruption of existing host protein complex formation to inhibit undesirable host responses. Due to the use of affinity precipitation approaches, the majority of studies have focused on how the virus takes advantage of the newly formed protein interactions for its own replication. Proximity labeling (PL), however, can also highlight transient and negative effects-those interactions which lead to dissociation from the existing protein complex. Here, we highlight the power of PL in combination with recombinant KSHV to study viral host interactions.


Asunto(s)
Biotinilación/métodos , Herpesvirus Humano 8/metabolismo , Factores Reguladores del Interferón/metabolismo , Proteómica , Sarcoma de Kaposi/virología , Proteínas Virales/metabolismo , Regulación Viral de la Expresión Génica , Células HEK293 , Interacciones Microbiota-Huesped , Humanos , Replicación Viral
8.
mBio ; 11(4)2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32843547

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) causes primary effusion lymphoma (PEL). The cellular transcription factor (TF) interferon (IFN) regulatory factor 4 (IRF4) is an essential oncogene in PEL, but its specific role in PEL and how KSHV deregulates IRF4 remain unknown. Here, we report that the KSHV latency protein viral interferon regulatory factor 3 (vIRF3) cooperates with IRF4 and cellular BATF (basic leucine zipper ATF-like TF) to drive a super-enhancer (SE)-mediated oncogenic transcriptional program in PEL. Chromatin immunoprecipitation coupled with next-generation sequencing (ChIP-Seq) experiments demonstrated that IRF4, vIRF3, and BATF cooccupy the SEs of key survival genes, in a pattern that is distinct from those seen with other IRF4-driven malignancies. All three proteins cooperatively drive SE-mediated IRF4 overexpression. Inactivation of vIRF3 and, to a lesser extent, BATF phenocopies the gene expression changes and loss of cellular viability observed upon inactivation of IRF4. In sum, this work suggests that KSHV vIRF3 and cellular IRF4 and BATF cooperate as oncogenic transcription factors on SEs to promote cellular survival and proliferation in KSHV-associated lymphomas.IMPORTANCE Kaposi's sarcoma-associated herpesvirus (KSHV) causes the aggressive disease primary effusion lymphoma (PEL). Here, we show that a viral transcription factor (vIRF3) cooperates with the cellular transcription factor IRF4 to control an oncogenic gene expression program in PEL cells. These proteins promote KSHV-mediated B cell transformation by activating the expression of prosurvival genes through super-enhancers. Our report thus demonstrates that this DNA tumor virus encodes a transcription factor that functions with cellular IRF4 to drive oncogenic transcriptional reprogramming.


Asunto(s)
Expresión Génica , Herpesvirus Humano 8/patogenicidad , Linfoma de Efusión Primaria/genética , Linfoma de Efusión Primaria/virología , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/virología , Línea Celular Tumoral , Humanos , Factores Reguladores del Interferón/genética , Proteínas Virales/genética , Latencia del Virus
9.
Virus Res ; 286: 198076, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32603670

RESUMEN

Ubiquitin Specific Protease 7 (USP7) is a deubiquitinating enzyme (DUB) that plays critical roles in the regulation of many cellular processes including epigenetics, tumour suppression, oncogenesis, DNA damage response, immunity and viral infection. USP7 was the first DUB associated with viral infection. Since then other DUB:viral protein interactions have been discovered, however, USP7 continues to be the most targeted DUB interacting with many proteins from various viruses. The selective pressures of evolution have allowed viruses to develop mechanisms that subvert host cellular machinery, promoting survival of the viral niche. Numerous viral proteins have been identified to target and usurp the function of USP7 to their advantage. This review explores novel developments in research focusing on the mechanisms underlying the manipulation of USP7 by viruses.


Asunto(s)
Peptidasa Específica de Ubiquitina 7/genética , Peptidasa Específica de Ubiquitina 7/metabolismo , Proteínas Virales/genética , Virus/genética , Animales , Inestabilidad Genómica , Humanos , Proteínas Inmediatas-Precoces , Ratones , Unión Proteica , Ubiquitina/metabolismo , Proteínas Virales/metabolismo , Virus/metabolismo
10.
New Microbes New Infect ; 32: 100604, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31719995

RESUMEN

Interactions between bacterial virulence and antimicrobial resistance are of increasing interest in clinical microbiology. On this account, antimicrobial resistance of Yersinia enterocolitica O:3 strains isolated from humans (n = 55), food-chain animals (n = 58) and companion animals (n = 13) was determined in relation to the absence or presence of the pYV plasmid-encoded virulence genes yadA and virF. There were no statistically significant associations between the rate of antimicrobial resistance and the presence or absence of the plasmid, in either human-derived or animal-derived strains. Therefore, it can be concluded that response to conventionally used antimicrobials in Y. enterocolitica O:3 strains is not dependent on pYV-encoded virulence determinants.

11.
J Virol ; 93(6)2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30626678

RESUMEN

Interferon (IFN) production and the subsequent induction of IFN-stimulated genes (ISGs) are highly effective innate strategies utilized by cells to protect against invading pathogens, including viruses. Critical components involved in this innate process are promyelocytic leukemia nuclear bodies (PML-NBs), which are subnuclear structures required for the development of a robust IFN response. As such, PML-NBs serve as an important hurdle for viruses to overcome to successfully establish an infection. Both Kaposi's sarcoma-associated herpesvirus (KSHV) and the closely related rhesus macaque rhadinovirus (RRV) are unique for encoding viral homologs of IFN regulatory factors (termed vIRFs) that can manipulate the host immune response by multiple mechanisms. All four KSHV vIRFs inhibit the induction of IFN, while vIRF1 and vIRF2 can inhibit ISG induction downstream of the IFN receptor. Less is known about the RRV vIRFs. RRV vIRF R6 can inhibit the induction of IFN by IRF3; however, it is not known whether any RRV vIRFs inhibit ISG induction following IFN receptor signaling. In our present study, we demonstrate that the RRV vIRF R12 aids viral replication in the presence of the type I IFN response. This is achieved in part through the disruption of PML-NBs and the inhibition of robust ISG transcription.IMPORTANCE KSHV and RRV encode a unique set of homologs of cellular IFN regulatory factors, termed vIRFs, which are hypothesized to help these viruses evade the innate immune response and establish infections in their respective hosts. Our work elucidates the role of one RRV vIRF, R12, and demonstrates that RRV can dampen the type I IFN response downstream of IFN signaling, which would be important for establishing a successful infection in vivo.


Asunto(s)
Factores Reguladores del Interferón/genética , Interferón Tipo I/genética , Cuerpos de Inclusión Intranucleares/genética , Leucemia Promielocítica Aguda/genética , Macaca mulatta/virología , Rhadinovirus/genética , Transducción de Señal/genética , Proteínas Virales/genética , Animales , Línea Celular , Herpesvirus Humano 8/genética , Humanos , Inmunidad Innata/genética , Factor 3 Regulador del Interferón/genética , Leucemia Promielocítica Aguda/virología , Receptores de Interferón/genética , Transcripción Genética/genética , Replicación Viral/genética
12.
Biochem Biophys Res Commun ; 496(4): 1128-1133, 2018 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-29397940

RESUMEN

Primary effusion lymphoma (PEL), strongly linked with latent infection of Kaposi's sarcoma-associated herpesvirus (KSHV), constitutively expresses cellular interferon regulatory factor 4 (IRF4) while suppressing the expression of B cell lymphoma 6 (BCL6). Recently, it was shown that IRF4, a key transcriptional repressor of BCL6, might be a pivotal regulator of KSHV for balancing between latency and its reactivation in PEL cells. However, the action of the BCL6-IRF4 transcription factor axis during KSHV's life cycle is not clear. Herein we found that the KSHV lytic protein viral interferon regulatory factor 4 (vIRF4) dramatically enhanced the transcriptional activity of the BCL6 through the inhibition of its negative regulator IRF4. Using a chromatin immunoprecipitation (ChIP) assay, we further showed that vIRF4 bound to the specific promoter region of IRF4, contributing to a dramatic suppression of IRF4 gene expression. Correspondingly, we also found BCL6 expression to be positively and inversely correlated with vIRF4 and IRF4 expression, respectively, during KSHV reactivation. Finally, we observed that these processes require efficient KSHV lytic replication. Thus, our findings suggest a crucial role of the BCL6-IRF4 axis in triggering the transition between KSHV latency and lytic reactivation.


Asunto(s)
Regulación Viral de la Expresión Génica/fisiología , Herpesvirus Humano 8/metabolismo , Factores Reguladores del Interferón/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Activación Transcripcional/fisiología , Proteínas Virales/metabolismo , Activación Viral/fisiología , Latencia del Virus/fisiología , Regulación hacia Abajo , Replicación Viral/fisiología
13.
Front Microbiol ; 8: 650, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28458662

RESUMEN

VirF is the master activator of virulence genes of Shigella and its expression is required for the invasion of the human intestinal mucosa by pathogenic bacteria. VirF was shown to directly activate the transcription of virB and icsA, which encode two essential proteins involved in the pathogenicity process, by binding their promoter regions. In this study, we demonstrate by band shift, enzymatic probing and cross-linking experiments that VirF, in addition to DNA, can also bind the icsA transcript and RnaG, an antisense non-coding small RNA that promotes the premature termination of icsA mRNA through a transcriptional attenuation mechanism. Furthermore, we show that VirF binds in vitro also other species of RNAs, although with lower specificity. The existence of VirF-RnaG and VirF-icsA mRNA complexes is confirmed in a pulldown assay carried out under experimental conditions that very close reproduce the in vivo conditions and that allows immobilized VirF to "fish" out RnaG and icsA mRNA from a total RNA extract. The VirF binding sites identified on both icsA mRNA and RnaG contain a 13 nucleotides stretch (5'-UUUUaGYcUuUau-3') that is the RNA-converted consensus sequence previously proposed for the VirF-DNA interaction. Band-shift assays with a synthetic RNA molecule whose sequence perfectly matches the consensus indicate that this signature plays a key role also in the VirF-RNA interaction, in particular when exposed in a stem-loop structure. To further explore the icsA-RnaG-VirF regulatory system, we developed an in vitro test (RNA-RNA Pairing Assay) in which pairing between icsA mRNA and synthetic RNAs that reproduce the individual stem-loop motifs of RnaG, was analyzed in the presence of VirF. This assay shows that this protein can prevent the formation of the kissing complex, defined as the initial nucleation points for RNA heteroduplex formation, between RnaG and icsA mRNA. Consistently, VirF alleviates the RnaG-mediated repression of icsA transcription in vitro. Therefore VirF, by hindering the icsA transcript-RnaG interaction, exhibits an activity opposed to that usually displayed by proteins, which generally assist the RNA-RNA interaction; this quite uncommon and new function and the regulatory implications of VirF as a potential RNA-binding protein are discussed.

14.
Biochem Biophys Res Commun ; 486(3): 700-705, 2017 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-28342865

RESUMEN

Before an infection can be completely established, the host immediately turns on the innate immune system through activating the interferon (IFN)-mediated antiviral pathway. Kaposi's sarcoma-associated herpesvirus (KSHV) utilizes a unique antagonistic mechanism of type I IFN-mediated host antiviral immunity by incorporating four viral interferon regulatory factors (vIRF1-4). Herein, we characterized novel immune evasion strategies of vIRF4 to inhibit the IRF7-mediated IFN-α production. KSHV vIRF4 specifically interacts with IRF7, resulting in inhibition of IRF7 dimerization and ultimately suppresses IRF7-mediated activation of type I IFN. These results suggest that each of the KSHV vIRFs, including vIRF4, subvert IFN-mediated anti-viral response via different mechanisms. Therefore, it is indicated that KSHV vIRFs are indeed a crucial immunomodulatory component of their life cycles.


Asunto(s)
Herpesvirus Humano 8/inmunología , Evasión Inmune , Factor 7 Regulador del Interferón/inmunología , Factores Reguladores del Interferón/inmunología , Interferón-alfa/inmunología , Proteínas Virales/inmunología , Regulación de la Expresión Génica , Genes Reporteros , Células HEK293 , Herpesvirus Humano 8/química , Humanos , Inmunidad Innata , Factor 7 Regulador del Interferón/genética , Factores Reguladores del Interferón/genética , Interferón-alfa/antagonistas & inhibidores , Interferón-alfa/genética , Luciferasas/genética , Luciferasas/metabolismo , Plásmidos/química , Plásmidos/metabolismo , Virus Sendai/genética , Virus Sendai/inmunología , Transducción de Señal , Transfección , Proteínas Virales/genética
15.
Front Mol Biosci ; 3: 61, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27747215

RESUMEN

Shigella is a highly adapted human pathogen, mainly found in the developing world and causing a severe enteric syndrome. The highly sophisticated infectious strategy of Shigella banks on the capacity to invade the intestinal epithelial barrier and cause its inflammatory destruction. The cellular pathogenesis and clinical presentation of shigellosis are the sum of the complex action of a large number of bacterial virulence factors mainly located on a large virulence plasmid (pINV). The expression of pINV genes is controlled by multiple environmental stimuli through a regulatory cascade involving proteins and sRNAs encoded by both the pINV and the chromosome. The primary regulator of the virulence phenotype is VirF, a DNA-binding protein belonging to the AraC family of transcriptional regulators. The virF gene, located on the pINV, is expressed only within the host, mainly in response to the temperature transition occurring when the bacterium transits from the outer environment to the intestinal milieu. VirF then acts as anti-H-NS protein and directly activates the icsA and virB genes, triggering the full expression of the invasion program of Shigella. In this review we will focus on the structure of VirF, on its sophisticated regulation, and on its role as major player in the path leading from the non-invasive to the invasive phenotype of Shigella. We will address also the involvement of VirF in mechanisms aimed at withstanding adverse conditions inside the host, indicating that this protein is emerging as a global regulator whose action is not limited to virulence systems. Finally, we will discuss recent observations conferring VirF the potential of a novel antibacterial target for shigellosis.

16.
Biochem Biophys Res Commun ; 479(4): 697-702, 2016 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-27693583

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is an etiological agent of Kaposi's sarcoma and primary effusion lymphoma. Like other herpesviruses, KSHV has two distinct life cycles: latent and lytic. Among KSHV latent genes, viral interferon regulatory factor 3 (vIRF3), which shares homology with cellular IRFs, is a multifunctional protein. To identify unknown functions of vIRF3, we performed luciferase-reporter assays in the presence of vIRF3. These analyses revealed that overexpression of vIRF3 inhibited T-cell factor (TCF)-dependent transcriptional activity. This TCF-dependent transcription was associated with the Wnt signaling pathway, which normally regulates embryonic development, but contributes to oncogenesis under dysregulated conditions. Using a mutagenesis analysis, we identified a CREB-binding protein-interaction motif (LXXLL) in vIRF3 as an important region for its inhibitory activity. Collectively, our findings provide insight into the dysregulation of host signaling pathways in KSHV-infected cells.


Asunto(s)
Proteína de Unión a CREB/química , Herpesvirus Humano 8/fisiología , Interacciones Huésped-Patógeno/genética , Factores Reguladores del Interferón/metabolismo , Factores de Transcripción TCF/antagonistas & inhibidores , Proteínas Virales/metabolismo , Secuencias de Aminoácidos , Núcleo Celular/metabolismo , Células HEK293 , Herpesvirus Humano 8/metabolismo , Humanos , Factores Reguladores del Interferón/química , Factores Reguladores del Interferón/genética , Mutación , Factores de Transcripción TCF/metabolismo , Transcripción Genética , Proteínas Virales/química , Proteínas Virales/genética , Latencia del Virus , Vía de Señalización Wnt , beta Catenina/metabolismo
17.
J Biol Chem ; 291(12): 6281-91, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26786098

RESUMEN

Viral interferon regulatory factor 1 (vIRF1), a Kaposi sarcoma herpesvirus protein, destabilizes p53 by inhibiting p53 acetylation and Hdm2 phosphorylation. This leads to increased ubiquitination and degradation of p53 by Hdm2, which cripples the cellular p53-mediated antiviral response. Ubiquitin-specific protease 7 (USP7) deubiquitinates p53 and Hdm2 and regulates their stability. We identified an EGPS consensus sequence in vIRF1, which is identical to that found in Epstein-Barr virus nuclear antigen 1 (EBNA1) that interacts with the N-terminal domain of USP7 (USP7-NTD). GST pulldown assays demonstrated that vIRF1 interacts with USP7-NTD via its EGPS motif. NMR heteronuclear single quantum correlation (HSQC) analysis revealed chemical perturbations after titration of USP7-NTD with vIRF1 (44)SPGEGPSGTG(53) peptide. In contrast, these perturbations were reduced with a mutant vIRF1 peptide, (44)SPGEGPAGTG(53). Fluorescence polarization analysis indicated that the vIRF1 peptide interacted with USP7-NTD with a Kd of 2.0 µm. The crystal structure of the USP7-NTD·vIRF1 peptide complex revealed an identical mode of binding as that of the EBNA1 peptide to USP7-NTD. We also showed that USP7 interacts with vIRF1 in U2OS cells. Decreased levels of p53, but not Hdm2 or ataxia telangiectasia-mutated (ATM), were seen after expression of vIRF1, but not with a vIRF1 mutant protein. Our results support a new role for vIRF1 through deregulation of the deubiquitinating enzyme USP7 to inhibit p53-mediated antiviral responses.


Asunto(s)
Herpesvirus Humano 8 , Factores Reguladores del Interferón/química , Ubiquitina Tiolesterasa/química , Proteínas Virales/química , Secuencias de Aminoácidos , Dominio Catalítico , Línea Celular Tumoral , Cristalografía por Rayos X , Humanos , Factores Reguladores del Interferón/fisiología , Modelos Moleculares , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina Tiolesterasa/fisiología , Peptidasa Específica de Ubiquitina 7 , Proteínas Virales/fisiología
18.
Biochem Biophys Res Commun ; 470(3): 650-656, 2016 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-26797279

RESUMEN

Kaposi's sarcoma-associated herpesvirus encodes several genes with sequence homology to cellular interferon regulatory factors. Among these, vIRF2 encoded by ORF K11.1 (short form) or K11 (full-length) participates in caspase-3-mediated inactivation of cellular IRF3 and slightly inhibits caspase-3 activity. Here, we have demonstrated that vIRF2 attenuates the transcriptional activity of forkhead box O3A protein via activation of the PI3K/Akt phosphatidylinositol 3-kinase/Akt pathway, inhibiting FOXO3A-mediated caspase-3 cleavage. Based on the collective findings, we suggest that vIRF2 acts as an activator in PI3K/Akt pathway.


Asunto(s)
Caspasa 3/metabolismo , Factores de Transcripción Forkhead/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Animales , Activación Enzimática , Proteína Forkhead Box O3 , Células HEK293 , Humanos , Factores Reguladores del Interferón , Ratones , Células 3T3 NIH , Regulación hacia Arriba/fisiología , Proteínas Virales
19.
Iran J Microbiol ; 7(4): 198-202, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26697158

RESUMEN

BACKGROUND AND OBJECTIVES: Enteroinvasive Escherichia coli (EIEC) is one the cause of acute diarrhea and bacillary dysentery in developing countries. Routine diagnostic microbiology tests are not capable to distinguish EIEC from other pathogenic or non-pathogenic E. coli. PCR, targeting ipaH, virF, virB and other virulence genes, is a diagnostic method for detecting E. coli pathotypes. Using PCR, we identified EIEC by PCR targeting ipaH and virF genes among E.coli isolates from patients with diarrhea at the selected hospitals in Tehran. MATERIALS AND METHODS: Isolates of E. coli were cultured from 140 specimens of patients with diarrhea using culture and IMViC test. DNA was extracted using commercial kits and and tested for uidA, ipaH and virF genes by PCR. RESULTS: Totally, 140 E. coli isolates were confirmed by IMViC tests and PCR targeting uidA gene. Of 140 E. coli isolates, 5 (3.6%) were positive for the ipaH gene, 4 (2.9%) contained virF gene and 4 (2.9%) were positive for both ipaH and virF genes. CONCLUSION: These results indicated that EIEC is a considerable acute diarrheagenic pathogen in adults and infants. Moreover, virF gene is suggested for evaluation of invasiveness of EIEC.

20.
Proc Natl Acad Sci U S A ; 112(31): E4306-15, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26199418

RESUMEN

Infection of cells with DNA viruses triggers innate immune responses mediated by DNA sensors. cGMP-AMP synthase (cGAS) is a key DNA sensor that produces the cyclic dinucleotide cGMP-AMP (cGAMP) upon activation, which binds to and activates stimulator of interferon genes (STING), leading to IFN production and an antiviral response. Kaposi's sarcoma-associated herpesvirus (KSHV) is a DNA virus that is linked to several human malignancies. We report that KSHV infection activates the cGAS-STING pathway, and that cGAS and STING also play an important role in regulating KSHV reactivation from latency. We screened KSHV proteins for their ability to inhibit this pathway and identified six viral proteins that block IFN-ß activation through this pathway. This study is the first report identifying multiple viral proteins encoded by a human DNA virus that inhibit the cGAS-STING DNA sensing pathway. One such protein, viral interferon regulatory factor 1 (vIRF1), targets STING by preventing it from interacting with TANK binding kinase 1 (TBK1), thereby inhibiting STING's phosphorylation and concomitant activation, resulting in an inhibition of the DNA sensing pathway. Our data provide a unique mechanism for the negative regulation of STING-mediated DNA sensing. Moreover, the depletion of vIRF1 in the context of KSHV infection prevented efficient viral reactivation and replication, and increased the host IFN response to KSHV. The vIRF1-expressing cells also inhibited IFN-ß production following infection with DNA pathogens. Collectively, our results demonstrate that gammaherpesviruses encode inhibitors that block cGAS-STING-mediated antiviral immunity, and that modulation of this pathway is important for viral transmission and the lifelong persistence of herpesviruses in the human population.


Asunto(s)
ADN Viral/metabolismo , Herpesvirus Humano 8/fisiología , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/metabolismo , Transducción de Señal , Secuencia de Bases , ADN Viral/genética , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Factor 1 Regulador del Interferón/metabolismo , Interferón beta/metabolismo , Datos de Secuencia Molecular , Motivos de Nucleótidos/genética , Sistemas de Lectura Abierta/genética , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Virales/metabolismo , Activación Viral , Latencia del Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA