Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Nature ; 619(7969): 338-347, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37380775

RESUMEN

Spillover events of avian influenza A viruses (IAVs) to humans could represent the first step in a future pandemic1. Several factors that limit the transmission and replication of avian IAVs in mammals have been identified. There are several gaps in our understanding to predict which virus lineages are more likely to cross the species barrier and cause disease in humans1. Here, we identified human BTN3A3 (butyrophilin subfamily 3 member A3)2 as a potent inhibitor of avian IAVs but not human IAVs. We determined that BTN3A3 is expressed in human airways and its antiviral activity evolved in primates. We show that BTN3A3 restriction acts primarily at the early stages of the virus life cycle by inhibiting avian IAV RNA replication. We identified residue 313 in the viral nucleoprotein (NP) as the genetic determinant of BTN3A3 sensitivity (313F or, rarely, 313L in avian viruses) or evasion (313Y or 313V in human viruses). However, avian IAV serotypes, such as H7 and H9, that spilled over into humans also evade BTN3A3 restriction. In these cases, BTN3A3 evasion is due to substitutions (N, H or Q) in NP residue 52 that is adjacent to residue 313 in the NP structure3. Thus, sensitivity or resistance to BTN3A3 is another factor to consider in the risk assessment of the zoonotic potential of avian influenza viruses.


Asunto(s)
Aves , Interacciones Microbiota-Huesped , Virus de la Influenza A , Gripe Aviar , Gripe Humana , Zoonosis Virales , Animales , Humanos , Aves/virología , Virus de la Influenza A/clasificación , Virus de la Influenza A/genética , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/transmisión , Gripe Aviar/virología , Gripe Humana/prevención & control , Gripe Humana/transmisión , Gripe Humana/virología , Primates , Sistema Respiratorio/metabolismo , Sistema Respiratorio/virología , Medición de Riesgo , Zoonosis Virales/prevención & control , Zoonosis Virales/transmisión , Zoonosis Virales/virología , Replicación Viral
2.
PLoS Pathog ; 20(8): e1012466, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39150989

RESUMEN

Most viral diseases display a variable clinical outcome due to differences in virus strain virulence and/or individual host susceptibility to infection. Understanding the biological mechanisms differentiating a viral infection displaying severe clinical manifestations from its milder forms can provide the intellectual framework toward therapies and early prognostic markers. This is especially true in arbovirus infections, where most clinical cases are present as mild febrile illness. Here, we used a naturally occurring vector-borne viral disease of ruminants, bluetongue, as an experimental system to uncover the fundamental mechanisms of virus-host interactions resulting in distinct clinical outcomes. As with most viral diseases, clinical symptoms in bluetongue can vary dramatically. We reproduced experimentally distinct clinical forms of bluetongue infection in sheep using three bluetongue virus (BTV) strains (BTV-1IT2006, BTV-1IT2013 and BTV-8FRA2017). Infected animals displayed clinical signs varying from clinically unapparent, to mild and severe disease. We collected and integrated clinical, haematological, virological, and histopathological data resulting in the analyses of 332 individual parameters from each infected and uninfected control animal. We subsequently used machine learning to select the key viral and host processes associated with disease pathogenesis. We identified and experimentally validated five different fundamental processes affecting the severity of bluetongue: (i) virus load and replication in target organs, (ii) modulation of the host type-I IFN response, (iii) pro-inflammatory responses, (iv) vascular damage, and (v) immunosuppression. Overall, we showed that an agnostic machine learning approach can be used to prioritise the different pathogenetic mechanisms affecting the disease outcome of an arbovirus infection.


Asunto(s)
Infecciones por Arbovirus , Virus de la Lengua Azul , Lengua Azul , Lengua Azul/virología , Lengua Azul/patología , Animales , Ovinos , Virus de la Lengua Azul/patogenicidad , Infecciones por Arbovirus/virología , Infecciones por Arbovirus/patología , Índice de Severidad de la Enfermedad , Modelos Animales de Enfermedad
3.
PLoS Biol ; 21(2): e3001941, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36757937

RESUMEN

Interactions between viruses during coinfections can influence viral fitness and population diversity, as seen in the generation of reassortant pandemic influenza A virus (IAV) strains. However, opportunities for interactions between closely related viruses are limited by a process known as superinfection exclusion (SIE), which blocks coinfection shortly after primary infection. Using IAVs, we asked whether SIE, an effect which occurs at the level of individual cells, could limit interactions between populations of viruses as they spread across multiple cells within a host. To address this, we first measured the kinetics of SIE in individual cells by infecting them sequentially with 2 isogenic IAVs, each encoding a different fluorophore. By varying the interval between addition of the 2 IAVs, we showed that early in infection SIE does not prevent coinfection, but that after this initial lag phase the potential for coinfection decreases exponentially. We then asked how the kinetics of SIE onset controlled coinfections as IAVs spread asynchronously across monolayers of cells. We observed that viruses at individual coinfected foci continued to coinfect cells as they spread, because all new infections were of cells that had not yet established SIE. In contrast, viruses spreading towards each other from separately infected foci could only establish minimal regions of coinfection before reaching cells where coinfection was blocked. This created a pattern of separate foci of infection, which was recapitulated in the lungs of infected mice, and which is likely to be applicable to many other viruses that induce SIE. We conclude that the kinetics of SIE onset segregate spreading viral infections into discrete regions, within which interactions between virus populations can occur freely, and between which they are blocked.


Asunto(s)
Coinfección , Gripe Humana , Orthomyxoviridae , Sobreinfección , Ratones , Animales , Humanos , Virus Reordenados
4.
PLoS Biol ; 19(12): e3001065, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34932557

RESUMEN

The pandemic spread of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the etiological agent of Coronavirus Disease 2019 (COVID-19), represents an ongoing international health crisis. A key symptom of SARS-CoV-2 infection is the onset of fever, with a hyperthermic temperature range of 38 to 41°C. Fever is an evolutionarily conserved host response to microbial infection that can influence the outcome of viral pathogenicity and regulation of host innate and adaptive immune responses. However, it remains to be determined what effect elevated temperature has on SARS-CoV-2 replication. Utilizing a three-dimensional (3D) air-liquid interface (ALI) model that closely mimics the natural tissue physiology of SARS-CoV-2 infection in the respiratory airway, we identify tissue temperature to play an important role in the regulation of SARS-CoV-2 infection. Respiratory tissue incubated at 40°C remained permissive to SARS-CoV-2 entry but refractory to viral transcription, leading to significantly reduced levels of viral RNA replication and apical shedding of infectious virus. We identify tissue temperature to play an important role in the differential regulation of epithelial host responses to SARS-CoV-2 infection that impact upon multiple pathways, including intracellular immune regulation, without disruption to general transcription or epithelium integrity. We present the first evidence that febrile temperatures associated with COVID-19 inhibit SARS-CoV-2 replication in respiratory epithelia. Our data identify an important role for tissue temperature in the epithelial restriction of SARS-CoV-2 independently of canonical interferon (IFN)-mediated antiviral immune defenses.


Asunto(s)
Células Epiteliales/inmunología , Calor , Inmunidad Innata/inmunología , Interferones/inmunología , Mucosa Respiratoria/inmunología , SARS-CoV-2/inmunología , Replicación Viral/inmunología , Adolescente , Animales , COVID-19/genética , COVID-19/inmunología , COVID-19/virología , Chlorocebus aethiops , Células Epiteliales/metabolismo , Células Epiteliales/virología , Femenino , Perfilación de la Expresión Génica/métodos , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/genética , Interferones/genética , Interferones/metabolismo , Masculino , Persona de Mediana Edad , Modelos Biológicos , RNA-Seq/métodos , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/virología , SARS-CoV-2/genética , SARS-CoV-2/fisiología , Técnicas de Cultivo de Tejidos , Células Vero , Replicación Viral/genética , Replicación Viral/fisiología
5.
EMBO Rep ; 22(9): e52547, 2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34197022

RESUMEN

Herpes simplex virus (HSV) establishes latent infection in long-lived neurons. During initial infection, neurons are exposed to multiple inflammatory cytokines but the effects of immune signaling on the nature of HSV latency are unknown. We show that initial infection of primary murine neurons in the presence of type I interferon (IFN) results in a form of latency that is restricted for reactivation. We also find that the subnuclear condensates, promyelocytic leukemia nuclear bodies (PML-NBs), are absent from primary sympathetic and sensory neurons but form with type I IFN treatment and persist even when IFN signaling resolves. HSV-1 genomes colocalize with PML-NBs throughout a latent infection of neurons only when type I IFN is present during initial infection. Depletion of PML prior to or following infection does not impact the establishment latency; however, it does rescue the ability of HSV to reactivate from IFN-treated neurons. This study demonstrates that viral genomes possess a memory of the IFN response during de novo infection, which results in differential subnuclear positioning and ultimately restricts the ability of genomes to reactivate.


Asunto(s)
Herpes Simple , Herpesvirus Humano 1 , Interferón Tipo I , Animales , Genoma Viral , Herpes Simple/genética , Herpesvirus Humano 1/genética , Interferón Tipo I/genética , Ratones , Latencia del Virus
6.
PLoS Pathog ; 16(12): e1009134, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33351855

RESUMEN

Mosquitoes are responsible for the transmission of many clinically important arboviruses that cause significant levels of annual mortality and socioeconomic health burden worldwide. Deciphering the mechanisms by which mosquitoes modulate arbovirus infection is crucial to understand how viral-host interactions promote vector transmission and human disease. SUMOylation is a post-translational modification that leads to the covalent attachment of the Small Ubiquitin-like MOdifier (SUMO) protein to host factors, which in turn can modulate their stability, interaction networks, sub-cellular localisation, and biochemical function. While the SUMOylation pathway is known to play a key role in the regulation of host immune defences to virus infection in humans, the importance of this pathway during arbovirus infection in mosquito vectors, such as Aedes aegypti (Ae. aegypti), remains unknown. Here we characterise the sequence, structure, biochemical properties, and tissue-specific expression profiles of component proteins of the Ae. aegypti SUMOylation pathway. We demonstrate significant biochemical differences between Ae. aegypti and Homo sapiens SUMOylation pathways and identify cell-type specific patterns of SUMO expression in Ae. aegypti tissues known to support arbovirus replication. Importantly, depletion of core SUMOylation effector proteins (SUMO, Ubc9 and PIAS) in Ae. aegypti cells led to enhanced levels of arbovirus replication from three different families; Zika (Flaviviridae), Semliki Forest (Togaviridae), and Bunyamwera (Bunyaviridae) viruses. Our findings identify an important role for mosquito SUMOylation in the cellular restriction of arboviruses that may directly influence vector competence and transmission of clinically important arboviruses.


Asunto(s)
Aedes/virología , Arbovirus/fisiología , Interacciones Huésped-Patógeno/fisiología , Mosquitos Vectores/virología , Replicación Viral/fisiología , Animales , Infecciones por Arbovirus/transmisión , Humanos , Sumoilación
7.
PLoS Pathog ; 15(3): e1007667, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30901352

RESUMEN

Host innate immune defences play a critical role in restricting the intracellular propagation and pathogenesis of invading viral pathogens. Here we show that the histone H3.3 chaperone HIRA (histone cell cycle regulator) associates with promyelocytic leukaemia nuclear bodies (PML-NBs) to stimulate the induction of innate immune defences against herpes simplex virus 1 (HSV-1) infection. Following the activation of innate immune signalling, HIRA localized at PML-NBs in a Janus-Associated Kinase (JAK), Cyclin Dependent Kinase (CDK), and Sp100-dependent manner. RNA-seq analysis revealed that HIRA promoted the transcriptional upregulation of a broad repertoire of host genes that regulate innate immunity to HSV-1 infection, including those involved in MHC-I antigen presentation, cytokine signalling, and interferon stimulated gene (ISG) expression. ChIP-seq analysis revealed that PML, the principle scaffolding protein of PML-NBs, was required for the enrichment of HIRA onto ISGs, identifying a role for PML in the HIRA-dependent regulation of innate immunity to virus infection. Our data identifies independent roles for HIRA in the intrinsic silencing of viral gene expression and the induction of innate immune defences to restrict the initiation and propagation of HSV-1 infection, respectively. These intracellular host defences are antagonized by the HSV-1 ubiquitin ligase ICP0, which disrupts the stable recruitment of HIRA to infecting viral genomes and PML-NBs at spatiotemporally distinct phases of infection. Our study highlights the importance of histone chaperones to regulate multiple phases of intracellular immunity to virus infection, findings that are likely to be highly pertinent in the cellular restriction of many clinically important viral pathogens.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Fibroblastos/inmunología , Infecciones por Herpesviridae/inmunología , Herpesvirus Humano 1/patogenicidad , Chaperonas de Histonas/metabolismo , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/inmunología , Factores de Transcripción/metabolismo , Proteínas de Ciclo Celular/genética , Células Cultivadas , Fibroblastos/citología , Fibroblastos/virología , Regulación Viral de la Expresión Génica , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/virología , Chaperonas de Histonas/genética , Humanos , Factores de Transcripción/genética , Replicación Viral
9.
PLoS Pathog ; 14(1): e1006769, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29309427

RESUMEN

Detection of viral nucleic acids plays a critical role in the induction of intracellular host immune defences. However, the temporal recruitment of immune regulators to infecting viral genomes remains poorly defined due to the technical difficulties associated with low genome copy-number detection. Here we utilize 5-Ethynyl-2'-deoxyuridine (EdU) labelling of herpes simplex virus 1 (HSV-1) DNA in combination with click chemistry to examine the sequential recruitment of host immune regulators to infecting viral genomes under low multiplicity of infection conditions. Following viral genome entry into the nucleus, PML-nuclear bodies (PML-NBs) rapidly entrapped viral DNA (vDNA) leading to a block in viral replication in the absence of the viral PML-NB antagonist ICP0. This pre-existing intrinsic host defence to infection occurred independently of the vDNA pathogen sensor IFI16 (Interferon Gamma Inducible Protein 16) and the induction of interferon stimulated gene (ISG) expression, demonstrating that vDNA entry into the nucleus alone is not sufficient to induce a robust innate immune response. Saturation of this pre-existing intrinsic host defence during HSV-1 ICP0-null mutant infection led to the stable recruitment of PML and IFI16 into vDNA complexes associated with ICP4, and led to the induction of ISG expression. This induced innate immune response occurred in a PML-, IFI16-, and Janus-Associated Kinase (JAK)-dependent manner and was restricted by phosphonoacetic acid, demonstrating that vDNA polymerase activity is required for the robust induction of ISG expression during HSV-1 infection. Our data identifies dual roles for PML in the sequential regulation of intrinsic and innate immunity to HSV-1 infection that are dependent on viral genome delivery to the nucleus and the onset of vDNA replication, respectively. These intracellular host defences are counteracted by ICP0, which targets PML for degradation from the outset of nuclear infection to promote vDNA release from PML-NBs and the onset of HSV-1 lytic replication.


Asunto(s)
Regulación Viral de la Expresión Génica/efectos de los fármacos , Herpes Simple/metabolismo , Herpesvirus Humano 1/fisiología , Interacciones Huésped-Patógeno , Cuerpos de Inclusión Viral/metabolismo , Proteína de la Leucemia Promielocítica/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Virales/metabolismo , Línea Celular , Línea Celular Transformada , Células Cultivadas , Química Clic , Eliminación de Gen , Herpes Simple/tratamiento farmacológico , Herpes Simple/patología , Herpes Simple/virología , Herpesvirus Humano 1/crecimiento & desarrollo , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Inmunidad Innata/efectos de los fármacos , Cuerpos de Inclusión Viral/efectos de los fármacos , Cuerpos de Inclusión Viral/patología , Cuerpos de Inclusión Viral/virología , Cinética , Lisogenia/efectos de los fármacos , Mutación , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteína de la Leucemia Promielocítica/antagonistas & inhibidores , Proteína de la Leucemia Promielocítica/genética , Interferencia de ARN , Inhibidores de la Transcriptasa Inversa/farmacología , Ubiquitina-Proteína Ligasas/genética , Proteínas Virales/genética , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
10.
Mol Cell ; 46(1): 79-90, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22405594

RESUMEN

Viral hijacking of cellular processes relies on the ability to mimic the structure or function of cellular proteins. Many viruses encode ubiquitin ligases to facilitate infection, although the mechanisms by which they select their substrates are often unknown. The Herpes Simplex Virus type-1-encoded E3 ubiquitin ligase, ICP0, promotes infection through degradation of cellular proteins, including the DNA damage response E3 ligases RNF8 and RNF168. Here we describe a mechanism by which this viral E3 hijacks a cellular phosphorylation-based targeting strategy to degrade RNF8. By mimicking a cellular phosphosite, ICP0 binds RNF8 via the RNF8 forkhead associated (FHA) domain. Phosphorylation of ICP0 T67 by CK1 recruits RNF8 for degradation and thereby promotes viral transcription, replication, and progeny production. We demonstrate that this mechanism may constitute a broader viral strategy to target other cellular factors, highlighting the importance of this region of the ICP0 protein in countering intrinsic antiviral defenses.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Herpesvirus Humano 1/fisiología , Proteínas Inmediatas-Precoces/metabolismo , Imitación Molecular/fisiología , Proteolisis , Ubiquitina-Proteína Ligasas/metabolismo , Replicación Viral/fisiología , Animales , Chlorocebus aethiops , Proteínas de Unión al ADN/genética , Células HeLa , Humanos , Proteínas Inmediatas-Precoces/genética , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , Transcripción Genética/fisiología , Ubiquitina-Proteína Ligasas/genética , Células Vero
11.
J Virol ; 90(9): 4807-4826, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26937035

RESUMEN

UNLABELLED: Small ubiquitin-like modifier (SUMO) is used by the intrinsic antiviral immune response to restrict viral pathogens, such as herpes simplex virus 1 (HSV-1). Despite characterization of the host factors that rely on SUMOylation to exert their antiviral effects, the enzymes that mediate these SUMOylation events remain to be defined. We show that unconjugated SUMO levels are largely maintained throughout infection regardless of the presence of ICP0, the HSV-1 SUMO-targeted ubiquitin ligase. Moreover, in the absence of ICP0, high-molecular-weight SUMO-conjugated proteins do not accumulate if HSV-1 DNA does not replicate. These data highlight the continued importance for SUMO signaling throughout infection. We show that the SUMO ligase protein inhibitor of activated STAT 4 (PIAS4) is upregulated during HSV-1 infection and localizes to nuclear domains that contain viral DNA. PIAS4 is recruited to sites associated with HSV-1 genome entry through SUMO interaction motif (SIM)-dependent mechanisms that are destabilized by ICP0. In contrast, PIAS4 accumulates in replication compartments through SIM-independent mechanisms irrespective of ICP0 expression. Depletion of PIAS4 enhances the replication of ICP0-null mutant HSV-1, which is susceptible to restriction by the intrinsic antiviral immune response. The mechanisms of PIAS4-mediated restriction are synergistic with the restriction mechanisms of a characterized intrinsic antiviral factor, promyelocytic leukemia protein, and are antagonized by ICP0. We provide the first evidence that PIAS4 is an intrinsic antiviral factor. This novel role for PIAS4 in intrinsic antiviral immunity contrasts with the known roles of PIAS proteins as suppressors of innate immunity. IMPORTANCE: Posttranslational modifications with small ubiquitin-like modifier (SUMO) proteins regulate multiple aspects of host immunity and viral replication. The protein inhibitor of activated STAT (PIAS) family of SUMO ligases is predominantly associated with the suppression of innate immune signaling. We now identify a unique and contrasting role for PIAS proteins as positive regulators of the intrinsic antiviral immune response to herpes simplex virus 1 (HSV-1) infection. We show that PIAS4 relocalizes to nuclear domains that contain viral DNA throughout infection. Depletion of PIAS4, either alone or in combination with the intrinsic antiviral factor promyelocytic leukemia protein, significantly impairs the intrinsic antiviral immune response to HSV-1 infection. Our data reveal a novel and dynamic role for PIAS4 in the cellular-mediated restriction of herpesviruses and establish a new functional role for the PIAS family of SUMO ligases in the intrinsic antiviral immune response to DNA virus infection.


Asunto(s)
Herpes Simple/genética , Herpes Simple/inmunología , Herpesvirus Humano 1/fisiología , Interacciones Huésped-Patógeno , Inmunidad Innata/genética , Proteínas Inhibidoras de STAT Activados/genética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Línea Celular , Replicación del ADN , ADN Viral , Progresión de la Enfermedad , Expresión Génica , Genoma Viral , Herpes Simple/metabolismo , Herpes Simple/virología , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Mutación , Proteínas Nucleares/metabolismo , Proteína de la Leucemia Promielocítica , Unión Proteica , Proteínas Inhibidoras de STAT Activados/química , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Proteínas Recombinantes de Fusión , Proteína SUMO-1/metabolismo , Sumoilación , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Replicación Viral
12.
J Virol ; 90(13): 5939-5952, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27099310

RESUMEN

UNLABELLED: Aspects of intrinsic antiviral immunity are mediated by promyelocytic leukemia nuclear body (PML-NB) constituent proteins. During herpesvirus infection, these antiviral proteins are independently recruited to nuclear domains that contain infecting viral genomes to cooperatively promote viral genome silencing. Central to the execution of this particular antiviral response is the small ubiquitin-like modifier (SUMO) signaling pathway. However, the participating SUMOylation enzymes are not fully characterized. We identify the SUMO ligase protein inhibitor of activated STAT1 (PIAS1) as a constituent PML-NB protein. We show that PIAS1 localizes at PML-NBs in a SUMO interaction motif (SIM)-dependent manner that requires SUMOylated or SUMOylation-competent PML. Following infection with herpes simplex virus 1 (HSV-1), PIAS1 is recruited to nuclear sites associated with viral genome entry in a SIM-dependent manner, consistent with the SIM-dependent recruitment mechanisms of other well-characterized PML-NB proteins. In contrast to that of Daxx and Sp100, however, the recruitment of PIAS1 is enhanced by PML. PIAS1 promotes the stable accumulation of SUMO1 at nuclear sites associated with HSV-1 genome entry, whereas the accumulation of other evaluated PML-NB proteins occurs independently of PIAS1. We show that PIAS1 cooperatively contributes to HSV-1 restriction through mechanisms that are additive to those of PML and cooperative with those of PIAS4. The antiviral mechanisms of PIAS1 are counteracted by ICP0, the HSV-1 SUMO-targeted ubiquitin ligase, which disrupts the recruitment of PIAS1 to nuclear domains that contain infecting HSV-1 genomes through mechanisms that do not directly result in PIAS1 degradation. IMPORTANCE: Adaptive, innate, and intrinsic immunity cooperatively and efficiently restrict the propagation of viral pathogens. Intrinsic immunity mediated by constitutively expressed cellular proteins represents the first line of intracellular defense against infection. PML-NB constituent proteins mediate aspects of intrinsic immunity to restrict herpes simplex virus 1 (HSV-1) as well as other viruses. These proteins repress viral replication through mechanisms that rely on SUMO signaling. However, the participating SUMOylation enzymes are not known. We identify the SUMO ligase PIAS1 as a constituent PML-NB antiviral protein. This finding distinguishes a SUMO ligase that may mediate signaling events important in PML-NB-mediated intrinsic immunity. Moreover, this research complements the recent identification of PIAS4 as an intrinsic antiviral factor, supporting a role for PIAS proteins as both positive and negative regulators of host immunity to virus infection.


Asunto(s)
Herpesvirus Humano 1/inmunología , Interacciones Huésped-Patógeno , Inmunidad Innata , Proteína de la Leucemia Promielocítica/química , Proteínas Inhibidoras de STAT Activados/metabolismo , Fibroblastos/virología , Prepucio , Regulación Viral de la Expresión Génica , Genoma Viral , Humanos , Masculino , Proteína de la Leucemia Promielocítica/metabolismo , Proteínas Inhibidoras de STAT Activados/genética , Factor de Transcripción STAT1/química , Factor de Transcripción STAT1/metabolismo , Proteína SUMO-1/antagonistas & inhibidores , Sumoilación , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Virales/metabolismo , Internalización del Virus , Replicación Viral
13.
Methods ; 90: 3-7, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25862948

RESUMEN

The ubiquitin-proteasome system is an essential cellular process that plays a fundamental role in the regulation of protein stability. This pathway is tightly controlled by a sequential cascade of enzymatic steps that culminates in the formation of a poly-ubiquitin chain onto the substrate protein targeted for 26S proteasome degradation. Through a process of co-evolution viruses have evolved mechanisms to utilize or suppress this pathway in order to enhance their replication and spread. One of the first proteins to be expressed during herpes simplex virus 1 (HSV-1) infection is ICP0, a viral RING-finger E3 ubiquitin ligase that targets a variety of cellular proteins for ubiquitination and proteasome-dependent degradation. This activity is required in order for ICP0 to efficiently stimulate the onset of HSV-1 lytic infection and viral reactivation from latency. While it is clear that the RING-finger domain of ICP0 plays an important role in the biology of HSV-1, methods for accurately quantifying its biochemical activity are currently lacking. Here we describe a protocol that enables the quantitative measurement of the ubiquitin ligase activity of ICP0 using near-infrared (IR) western blot imaging. The use of such imaging technology provides an accurate means to examine the biochemical and kinetic parameters of RING-finger ubiquitin ligases in solution, and may provide significant application for inhibitor studies.


Asunto(s)
Western Blotting/métodos , Herpesvirus Humano 1/enzimología , Proteínas Inmediatas-Precoces/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Herpesvirus Humano 1/patogenicidad , Ubiquitinación
14.
J Gen Virol ; 96(11): 3236-3242, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26361997

RESUMEN

Chronic infection of the liver by hepatitis C virus (HCV) induces a range of host factors including IFN-stimulated genes such as ISG15. ISG15 functions as an antiviral factor that limits virus replication. Previous studies have suggested that ISG15 could influence HCV replication in both a positive and a negative manner. In this report, we determined the effect of ISG15 on HCV RNA replication in two independent cell lines that support viral genome synthesis by inhibiting ISG15 expression through small interfering RNA, short-hairpin RNA and CRISPR/Cas9 gene knockout approaches. Our results demonstrated that ISG15 impairs HCV RNA replication in both the presence and absence of IFN stimulation, consistent with an antiviral role for ISG15 during HCV infection. ISG15 conjugation to protein substrates typically requires the E3 ligase, HERC5. Our results showed that the inhibitory effect of ISG15 on HCV RNA replication does not require its conjugation to substrates by HERC5.


Asunto(s)
Citocinas/inmunología , Hepacivirus/genética , Hepatitis C/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , ARN Viral/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinas/inmunología , Citocinas/genética , Hepacivirus/fisiología , Hepatitis C/genética , Hepatitis C/virología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , ARN Viral/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinas/genética , Replicación Viral
15.
J Virol ; 88(5): 2763-74, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24352468

RESUMEN

UNLABELLED: Herpes simplex virus type 1 immediate-early protein ICP0 is an E3 ubiquitin ligase of the RING finger class that degrades several cellular proteins during infection. This activity is essential for its functions in stimulating efficient lytic infection and productive reactivation from latency. ICP0 targets a number of proteins that are modified by the small ubiquitin-like SUMO family of proteins, and it includes a number of short sequences that are related to SUMO interaction motifs (SIMs). Therefore, ICP0 has characteristics that are related to those of cellular SUMO-targeted ubiquitin ligase enzymes. Here, we analyze the impact of mutation of a number of SIM-like sequences (SLSs) within ICP0 on HSV-1 replication and gene expression and their requirement for ICP0-mediated degradation of both sumoylated and unmodified promyelocytic leukemia (PML) and other sumoylated cellular proteins. One SLS in the central portion of the ICP0 sequence (SLS4) was found to be absolutely required for targeting cellular sumoylated species in general and sumoylated forms of PML other than those of PML isoform I. Mutation of a group of SLSs in the C-terminal quarter of ICP0 also reduced ICP0-mediated degradation of sumoylated PML in a cooperative manner. Although mutation of individual SLSs caused only modest decreases in viral replication, combined mutation of SLS4 with SLS sequences in the C-terminal quarter of the protein reduced plaque formation efficiency by up to two orders of magnitude. These results provide further evidence that the biological activities of ICP0 are connected with host cell sumoylation events. IMPORTANCE: Herpes simplex virus type 1 protein ICP0 plays important roles in regulating the initial stages of lytic infection and productive reactivation from latency. ICP0 mediates its effects through inducing the degradation of cellular proteins that have repressive effects on viral gene expression. An increasing number of cellular proteins are known to be sensitive to ICP0-mediated degradation; therefore, it is important to understand how ICP0 selects its substrates for degradation. This study identifies sequence motifs within ICP0 that are involved in targeting cellular proteins that are modified by the SUMO family of ubiquitin-like proteins and describes how mutation of combinations of these motifs causes a 100-fold defect in viral infectivity.


Asunto(s)
Herpesvirus Humano 1/fisiología , Proteínas Inmediatas-Precoces/metabolismo , Dominios y Motivos de Interacción de Proteínas , Proteína SUMO-1/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Línea Celular , Regulación Viral de la Expresión Génica , Genoma Viral , Humanos , Proteínas Inmediatas-Precoces/química , Proteínas Inmediatas-Precoces/genética , Espacio Intracelular/metabolismo , Datos de Secuencia Molecular , Mutación , Proteínas Nucleares/metabolismo , Proteína de la Leucemia Promielocítica , Unión Proteica , Transporte de Proteínas , Proteolisis , Dominios RING Finger , Alineación de Secuencia , Sumoilación , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Ensayo de Placa Viral , Replicación Viral
16.
J Virol ; 88(14): 7738-52, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24760893

RESUMEN

Myxovirus resistance 2 (Mx2/MxB) has recently been uncovered as an effector of the anti-HIV-1 activity of type I interferons (IFNs) that inhibits HIV-1 at an early stage postinfection, after reverse transcription but prior to proviral integration into host DNA. The mechanistic details of Mx2 antiviral activity are not yet understood, but a few substitutions in the HIV-1 capsid have been shown to confer resistance to Mx2. Through a combination of in vitro evolution and unbiased mutagenesis, we further map the determinants of sensitivity to Mx2 and reveal that multiple capsid (CA) surfaces define sensitivity to Mx2. Intriguingly, we reveal an unanticipated sensitivity determinant within the C-terminal domain of capsid. We also report that Mx2s derived from multiple primate species share the capacity to potently inhibit HIV-1, whereas selected nonprimate orthologs have no such activity. Like TRIM5α, another CA targeting antiretroviral protein, primate Mx2s exhibit species-dependent variation in antiviral specificity against at least one extant virus and multiple HIV-1 capsid mutants. Using a combination of chimeric Mx2 proteins and evolution-guided approaches, we reveal that a single residue close to the N terminus that has evolved under positive selection can determine antiviral specificity. Thus, the variable N-terminal region can define the spectrum of viruses inhibited by Mx2. Importance: Type I interferons (IFNs) inhibit the replication of most mammalian viruses. IFN stimulation upregulates hundreds of different IFN-stimulated genes (ISGs), but it is often unclear which ISGs are responsible for inhibition of a given virus. Recently, Mx2 was identified as an ISG that contributes to the inhibition of HIV-1 replication by type I IFN. Thus, Mx2 might inhibit HIV-1 replication in patients, and this inhibitory action might have therapeutic potential. The mechanistic details of how Mx2 inhibits HIV-1 are currently unclear, but the HIV-1 capsid protein is the likely viral target. Here, we determine the regions of capsid that specify sensitivity to Mx2. We demonstrate that Mx2 from multiple primates can inhibit HIV-1, whereas Mx2 from other mammals (dogs and sheep) cannot. We also show that primate variants of Mx2 differ in the spectrum of lentiviruses they inhibit and that a single residue in Mx2 can determine this antiviral specificity.


Asunto(s)
Proteína p24 del Núcleo del VIH/inmunología , VIH-1/inmunología , Proteínas de Resistencia a Mixovirus/inmunología , Animales , Análisis Mutacional de ADN , Evolución Molecular , Proteína p24 del Núcleo del VIH/genética , VIH-1/genética , Humanos , Mutagénesis
17.
PLoS Pathog ; 9(8): e1003514, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23950709

RESUMEN

Herpes simplex virus type 1 (HSV-1) is a neurotropic virus causing vesicular oral or genital skin lesions, meningitis and other diseases particularly harmful in immunocompromised individuals. To comprehensively investigate the complex interaction between HSV-1 and its host we combined two genome-scale screens for host factors (HFs) involved in virus replication. A yeast two-hybrid screen for protein interactions and a RNA interference (RNAi) screen with a druggable genome small interfering RNA (siRNA) library confirmed existing and identified novel HFs which functionally influence HSV-1 infection. Bioinformatic analyses found the 358 HFs were enriched for several pathways and multi-protein complexes. Of particular interest was the identification of Med23 as a strongly anti-viral component of the largely pro-viral Mediator complex, which links specific transcription factors to RNA polymerase II. The anti-viral effect of Med23 on HSV-1 replication was confirmed in gain-of-function gene overexpression experiments, and this inhibitory effect was specific to HSV-1, as a range of other viruses including Vaccinia virus and Semliki Forest virus were unaffected by Med23 depletion. We found Med23 significantly upregulated expression of the type III interferon family (IFN-λ) at the mRNA and protein level by directly interacting with the transcription factor IRF7. The synergistic effect of Med23 and IRF7 on IFN-λ induction suggests this is the major transcription factor for IFN-λ expression. Genotypic analysis of patients suffering recurrent orofacial HSV-1 outbreaks, previously shown to be deficient in IFN-λ secretion, found a significant correlation with a single nucleotide polymorphism in the IFN-λ3 (IL28b) promoter strongly linked to Hepatitis C disease and treatment outcome. This paper describes a link between Med23 and IFN-λ, provides evidence for the crucial role of IFN-λ in HSV-1 immune control, and highlights the power of integrative genome-scale approaches to identify HFs critical for disease progression and outcome.


Asunto(s)
Genoma Humano , Herpesvirus Humano 1/fisiología , Interleucinas/biosíntesis , Complejo Mediador/biosíntesis , Regulación hacia Arriba , Replicación Viral/fisiología , Eliminación de Gen , Células HeLa , Herpes Simple/genética , Herpes Simple/inmunología , Herpes Simple/metabolismo , Humanos , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/inmunología , Factor 7 Regulador del Interferón/metabolismo , Interferones , Interleucinas/genética , Interleucinas/inmunología , Complejo Mediador/genética , Complejo Mediador/inmunología , Polimorfismo de Nucleótido Simple , ARN Polimerasa II/genética , ARN Polimerasa II/inmunología , ARN Polimerasa II/metabolismo
18.
Nature ; 462(7275): 886-90, 2009 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-20016594

RESUMEN

Mutations in BRCA1 are associated with a high risk of breast and ovarian cancer. BRCA1 participates in the DNA damage response and acts as a ubiquitin ligase. However, its regulation remains poorly understood. Here we report that BRCA1 is modified by small ubiquitin-like modifier (SUMO) in response to genotoxic stress, and co-localizes at sites of DNA damage with SUMO1, SUMO2/3 and the SUMO-conjugating enzyme Ubc9. PIAS SUMO E3 ligases co-localize with and modulate SUMO modification of BRCA1, and are required for BRCA1 ubiquitin ligase activity in cells. In vitro SUMO modification of the BRCA1/BARD1 heterodimer greatly increases its ligase activity, identifying it as a SUMO-regulated ubiquitin ligase (SRUbL). Further, PIAS SUMO ligases are required for complete accumulation of double-stranded DNA (dsDNA) damage-repair proteins subsequent to RNF8 accrual, and for proficient double-strand break repair. These data demonstrate that the SUMOylation pathway plays a significant role in mammalian DNA damage response.


Asunto(s)
Proteína BRCA1/metabolismo , Daño del ADN , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Roturas del ADN de Doble Cadena , Reparación del ADN , Células HeLa , Histonas/metabolismo , Humanos , Proteínas Inhibidoras de STAT Activados/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
19.
EMBO J ; 29(5): 943-55, 2010 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-20075863

RESUMEN

The ICP0 protein of herpes simplex virus type 1 is an E3 ubiquitin ligase and transactivator required for the efficient switch between latent and lytic infection. As DNA damaging treatments are known to reactivate latent virus, we wished to explore whether ICP0 modulates the cellular response to DNA damage. We report that ICP0 prevents accumulation of repair factors at cellular damage sites, acting between recruitment of the mediator proteins Mdc1 and 53BP1. We identify RNF8 and RNF168, cellular histone ubiquitin ligases responsible for anchoring repair factors at sites of damage, as new targets for ICP0-mediated degradation. By targeting these ligases, ICP0 expression results in loss of ubiquitinated forms of H2A, mobilization of DNA repair proteins and enhanced viral fitness. Our study raises the possibility that the ICP0-mediated control of histone ubiquitination may link DNA repair, relief of transcriptional repression, and activation of latent viral genomes.


Asunto(s)
Reparación del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Herpesvirus Humano 1/metabolismo , Histonas/metabolismo , Proteínas Inmediatas-Precoces/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/fisiología , Animales , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Daño del ADN/genética , Daño del ADN/fisiología , Reparación del ADN/genética , Recuperación de Fluorescencia tras Fotoblanqueo , Técnica del Anticuerpo Fluorescente , Células HeLa , Herpesvirus Humano 1/crecimiento & desarrollo , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Immunoblotting , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/genética , Ubiquitinación/fisiología , Células Vero
20.
bioRxiv ; 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39185184

RESUMEN

Herpesviruses are ubiquitous pathogens that cause a wide range of disease. Upon nuclear entry, their genomes associate with histones and chromatin modifying enzymes that regulate the progression of viral transcription and outcome of infection. While the composition and modification of viral chromatin has been extensively studied on bulk populations of infected cells by chromatin immunoprecipitation, this key regulatory process remains poorly defined at single-genome resolution. Here we use high-resolution quantitative imaging to investigate the spatial proximity of canonical and variant histones at individual Herpes Simplex Virus 1 (HSV-1) genomes within the first 90 minutes of infection. We identify significant population heterogeneity in the stable enrichment and spatial proximity of canonical histones (H2A, H2B, H3.1) at viral DNA (vDNA) relative to established promyelocytic leukaemia nuclear body (PML-NB) host factors that are actively recruited to viral genomes upon nuclear entry. We show the replication-independent histone H3.3/H4 chaperone Daxx to cooperate with PML to mediate the enrichment and spatial localization of variant histone H3.3 at vDNA that limits the rate of HSV-1 genome decompaction to restrict the progress of immediate-early (IE) transcription. This host response is counteracted by the viral ubiquitin ligase ICP0, which degrades PML to disperse Daxx and variant histone H3.3 from vDNA to stimulate the progression of viral genome expansion, IE transcription, and onset of HSV-1 replication. Our data support a model of intermediate and sequential histone assembly initiated by Daxx that limits the rate of HSV-1 genome decompaction independently of the stable enrichment of histones H2A and H2B at vDNA required to facilitate canonical nucleosome assembly. We identify HSV-1 genome decompaction upon nuclear infection to play a key role in the initiation and functional outcome of HSV-1 lytic infection, findings pertinent to the transcriptional regulation of many nuclear replicating herpesvirus pathogens.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA