Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(13): 3410-3425.e17, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34062120

RESUMEN

To control viral infection, vertebrates rely on both inducible interferon responses and less well-characterized cell-intrinsic responses composed of "at the ready" antiviral effector proteins. Here, we show that E3 ubiquitin ligase TRIM7 is a cell-intrinsic antiviral effector that restricts multiple human enteroviruses by targeting viral 2BC, a membrane remodeling protein, for ubiquitination and proteasome-dependent degradation. Selective pressure exerted by TRIM7 results in emergence of a TRIM7-resistant coxsackievirus with a single point mutation in the viral 2C ATPase/helicase. In cultured cells, the mutation helps the virus evade TRIM7 but impairs optimal viral replication, and this correlates with a hyperactive and structurally plastic 2C ATPase. Unexpectedly, the TRIM7-resistant virus has a replication advantage in mice and causes lethal pancreatitis. These findings reveal a unique mechanism for targeting enterovirus replication and provide molecular insight into the benefits and trade-offs of viral evolution imposed by a host restriction factor.


Asunto(s)
Enterovirus/fisiología , Enterovirus/patogenicidad , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Replicación Viral/fisiología , Adenosina Trifosfatasas/metabolismo , Animales , Línea Celular , Femenino , Humanos , Inflamación/patología , Ratones Endogámicos C57BL , Mutación/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Proteolisis , ARN Viral/metabolismo , Ubiquitina/metabolismo , Proteínas Virales/genética
2.
Nat Immunol ; 20(4): 493-502, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30833792

RESUMEN

Interferon-stimulated genes (ISGs) form the backbone of the innate immune system and are important for limiting intra- and intercellular viral replication and spread. We conducted a mass-spectrometry-based survey to understand the fundamental organization of the innate immune system and to explore the molecular functions of individual ISGs. We identified interactions between 104 ISGs and 1,401 cellular binding partners engaging in 2,734 high-confidence interactions. 90% of these interactions are unreported so far, and our survey therefore illuminates a far wider activity spectrum of ISGs than is currently known. Integration of the resulting ISG-interaction network with published datasets and functional studies allowed us to identify regulators of immunity and processes related to the immune system. Given the extraordinary robustness of the innate immune system, this ISG network may serve as a blueprint for therapeutic targeting of cellular systems to efficiently fight viral infections.


Asunto(s)
Inmunidad Innata , Interferones/fisiología , Mapeo de Interacción de Proteínas , Antígenos de Neoplasias/metabolismo , Biomarcadores de Tumor/metabolismo , Proteínas Portadoras/metabolismo , Línea Celular , Expresión Génica , Glicoproteínas/metabolismo , Células HEK293 , Células HeLa , Humanos , Inmunidad Innata/genética , Espectrometría de Masas , Receptores CCR4/metabolismo , Receptores de Péptidos/metabolismo , Ribonucleoproteínas/metabolismo , Proteínas Virales/metabolismo
3.
Immunity ; 50(4): 907-923, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30995506

RESUMEN

Type I interferons (IFNs) (IFN-α, IFN-ß) and type III IFNs (IFN-λ) share many properties, including induction by viral infection, activation of shared signaling pathways, and transcriptional programs. However, recent discoveries have revealed context-specific functional differences. Here, we provide a comprehensive review of type I and type III IFN activities, highlighting shared and distinct features from molecular mechanisms through physiological responses. Beyond discussing canonical antiviral functions, we consider the adaptive immune priming, anti-tumor, and autoimmune functions of IFNs. We discuss a model wherein type III IFNs serve as a front-line defense that controls infection at epithelial barriers while minimizing damaging inflammatory responses, reserving the more potent type I IFN response for when local responses are insufficient. In this context, we discuss current therapeutic applications targeting these cytokine pathways and highlight gaps in understanding of the biology of type I and type III IFNs in health and disease.


Asunto(s)
Interferón Tipo I/inmunología , Interferones/inmunología , Inmunidad Adaptativa , Animales , Antivirales/uso terapéutico , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/inmunología , Células Epiteliales/inmunología , Femenino , Humanos , Interferón Tipo I/efectos adversos , Interferón Tipo I/uso terapéutico , Interferones/efectos adversos , Interferones/uso terapéutico , Masculino , Intercambio Materno-Fetal/inmunología , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Especificidad de Órganos , Embarazo , Transducción de Señal/inmunología , Transcripción Genética , Transcriptoma , Virosis/tratamiento farmacológico , Virosis/inmunología , Interferón lambda
4.
Nature ; 609(7928): 793-800, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35944563

RESUMEN

The RNA genome of SARS-CoV-2 contains a 5' cap that facilitates the translation of viral proteins, protection from exonucleases and evasion of the host immune response1-4. How this cap is made in SARS-CoV-2 is not completely understood. Here we reconstitute the N7- and 2'-O-methylated SARS-CoV-2 RNA cap (7MeGpppA2'-O-Me) using virally encoded non-structural proteins (nsps). We show that the kinase-like nidovirus RdRp-associated nucleotidyltransferase (NiRAN) domain5 of nsp12 transfers the RNA to the amino terminus of nsp9, forming a covalent RNA-protein intermediate (a process termed RNAylation). Subsequently, the NiRAN domain transfers the RNA to GDP, forming the core cap structure GpppA-RNA. The nsp146 and nsp167 methyltransferases then add methyl groups to form functional cap structures. Structural analyses of the replication-transcription complex bound to nsp9 identified key interactions that mediate the capping reaction. Furthermore, we demonstrate in a reverse genetics system8 that the N terminus of nsp9 and the kinase-like active-site residues in the NiRAN domain are required for successful SARS-CoV-2 replication. Collectively, our results reveal an unconventional mechanism by which SARS-CoV-2 caps its RNA genome, thus exposing a new target in the development of antivirals to treat COVID-19.


Asunto(s)
Caperuzas de ARN , ARN Viral , SARS-CoV-2 , Proteínas Virales , Antivirales , COVID-19/virología , Dominio Catalítico , Guanosina Difosfato/metabolismo , Humanos , Metiltransferasas/metabolismo , Nucleotidiltransferasas/química , Nucleotidiltransferasas/metabolismo , Dominios Proteicos , Caperuzas de ARN/química , Caperuzas de ARN/genética , Caperuzas de ARN/metabolismo , ARN Viral/química , ARN Viral/genética , ARN Viral/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , SARS-CoV-2/enzimología , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Tratamiento Farmacológico de COVID-19
5.
Nature ; 589(7842): 456-461, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33328639

RESUMEN

Autophagy, a process of degradation that occurs via the lysosomal pathway, has an essential role in multiple aspects of immunity, including immune system development, regulation of innate and adaptive immune and inflammatory responses, selective degradation of intracellular microorganisms, and host protection against infectious diseases1,2. Autophagy is known to be induced by stimuli such as nutrient deprivation and suppression of mTOR, but little is known about how autophagosomal biogenesis is initiated in mammalian cells in response to viral infection. Here, using genome-wide short interfering RNA screens, we find that the endosomal protein sorting nexin 5 (SNX5)3,4 is essential for virus-induced, but not for basal, stress- or endosome-induced, autophagy. We show that SNX5 deletion increases cellular susceptibility to viral infection in vitro, and that Snx5 knockout in mice enhances lethality after infection with several human viruses. Mechanistically, SNX5 interacts with beclin 1 and ATG14-containing class III phosphatidylinositol-3-kinase (PI3KC3) complex 1 (PI3KC3-C1), increases the lipid kinase activity of purified PI3KC3-C1, and is required for endosomal generation of phosphatidylinositol-3-phosphate (PtdIns(3)P) and recruitment of the PtdIns(3)P-binding protein WIPI2 to virion-containing endosomes. These findings identify a context- and organelle-specific mechanism-SNX5-dependent PI3KC3-C1 activation at endosomes-for initiation of autophagy during viral infection.


Asunto(s)
Autofagia/inmunología , Nexinas de Clasificación/metabolismo , Virus/inmunología , Animales , Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Beclina-1/metabolismo , Línea Celular , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Endosomas/metabolismo , Femenino , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño/genética , Nexinas de Clasificación/deficiencia , Nexinas de Clasificación/genética , Proteínas de Transporte Vesicular/metabolismo
6.
PLoS Pathog ; 19(11): e1011719, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37939149

RESUMEN

Clinical studies report that viral infections promote acute or chronic bacterial infections at multiple host sites. These viral-bacterial co-infections are widely linked to more severe clinical outcomes. In experimental models in vitro and in vivo, virus-induced interferon responses can augment host susceptibility to secondary bacterial infection. Here, we used a cell-based screen to assess 389 interferon-stimulated genes (ISGs) for their ability to induce chronic Pseudomonas aeruginosa infection. We identified and validated five ISGs that were sufficient to promote bacterial infection. Furthermore, we dissected the mechanism of action of hexokinase 2 (HK2), a gene involved in the induction of aerobic glycolysis, commonly known as the Warburg effect. We report that HK2 upregulation mediates the induction of Warburg effect and secretion of L-lactate, which enhances chronic P. aeruginosa infection. These findings elucidate how the antiviral immune response renders the host susceptible to secondary bacterial infection, revealing potential strategies for viral-bacterial co-infection treatment.


Asunto(s)
Infecciones Bacterianas , Coinfección , Virosis , Virus , Humanos , Interferones/metabolismo , Virus/metabolismo
7.
EMBO Rep ; 24(9): e56901, 2023 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-37497756

RESUMEN

Interferons control viral infection by inducing the expression of antiviral effector proteins encoded by interferon-stimulated genes (ISGs). The field has mostly focused on identifying individual antiviral ISG effectors and defining their mechanisms of action. However, fundamental gaps in knowledge about the interferon response remain. For example, it is not known how many ISGs are required to protect cells from a particular virus, though it is theorized that numerous ISGs act in concert to achieve viral inhibition. Here, we used CRISPR-based loss-of-function screens to identify a markedly limited set of ISGs that confer interferon-mediated suppression of a model alphavirus, Venezuelan equine encephalitis virus (VEEV). We show via combinatorial gene targeting that three antiviral effectors-ZAP, IFIT3, and IFIT1-together constitute the majority of interferon-mediated restriction of VEEV, while accounting for < 0.5% of the interferon-induced transcriptome. Together, our data suggest a refined model of the antiviral interferon response in which a small subset of "dominant" ISGs may confer the bulk of the inhibition of a given virus.


Asunto(s)
Virus de la Encefalitis Equina Venezolana , Virus , Animales , Caballos , Interferones , Línea Celular , Replicación Viral , Antivirales/farmacología , Virus de la Encefalitis Equina Venezolana/fisiología
8.
Nature ; 568(7751): 249-253, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30894749

RESUMEN

The non-canonical NF-κB signalling cascade is essential for lymphoid organogenesis, B cell maturation, osteoclast differentiation, and inflammation in mammals1,2; dysfunction of this system is associated with human diseases, including immunological disorders and cancer3-6. Although expression of NF-κB-inducing kinase (NIK, also known as MAP3K14) is the rate-limiting step in non-canonical NF-κB pathway activation2,7, the mechanisms by which transcriptional responses are regulated remain largely unknown. Here we show that the sine oculis homeobox (SIX) homologue family transcription factors SIX1 and SIX2 are integral components of the non-canonical NF-κB signalling cascade. The developmentally silenced SIX proteins are reactivated in differentiated macrophages by NIK-mediated suppression of the ubiquitin proteasome pathway. Consequently, SIX1 and SIX2 target a subset of inflammatory gene promoters and directly inhibit the trans-activation function of the transcription factors RELA and RELB in a negative feedback circuit. In support of a physiologically pivotal role for SIX proteins in host immunity, a human SIX1 transgene suppressed inflammation and promoted the recovery of mice from endotoxic shock. In addition, SIX1 and SIX2 protected RAS/P53-driven non-small-cell lung carcinomas from inflammatory cell death induced by SMAC-mimetic chemotherapeutic agents (small-molecule activators of the non-canonical NF-κB pathway). Our findings identify a NIK-SIX signalling axis that fine-tunes inflammatory gene expression programs under both physiological and pathological conditions.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Inflamación/metabolismo , FN-kappa B/deficiencia , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Animales , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Femenino , Fibroblastos , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Proteínas de Homeodominio/inmunología , Humanos , Inflamación/genética , Listeria monocytogenes/inmunología , Masculino , Ratones , FN-kappa B/genética , Proteínas del Tejido Nervioso/inmunología , Regiones Promotoras Genéticas , Shigella flexneri/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIB/metabolismo , Quinasa de Factor Nuclear kappa B
9.
Nat Methods ; 18(12): 1542-1551, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34824475

RESUMEN

Transneuronal viruses are powerful tools for tracing neuronal circuits or delivering genes to specific neurons in the brain. While there are multiple retrograde viruses, few anterograde viruses are available. Further, available anterograde viruses often have limitations such as retrograde transport, high neuronal toxicity or weak signals. We developed an anterograde viral system based on a live attenuated vaccine for yellow fever-YFV-17D. Replication- or packaging-deficient mutants of YFV-17D can be reconstituted in the brain, leading to efficient synapse-specific and anterograde-only transneuronal spreading, which can be controlled to achieve either monosynaptic or polysynaptic tracing. Moreover, inducible transient replication of YFV-17D mutant is sufficient to induce permanent transneuronal genetic modifications without causing neuronal toxicity. The engineered YFV-17D systems can be used to express fluorescent markers, sensors or effectors in downstream neurons, thus providing versatile tools for mapping and functionally controlling neuronal circuits.


Asunto(s)
Desarrollo de Vacunas , Vacuna contra la Fiebre Amarilla/inmunología , Fiebre Amarilla/inmunología , Fiebre Amarilla/prevención & control , Animales , Anticuerpos Antivirales/inmunología , Encéfalo/patología , Dependovirus , Electrofisiología , Colorantes Fluorescentes , Células HEK293 , Humanos , Ratones , Mutación , Neuronas/patología , Sistemas de Lectura Abierta , Vacunas Atenuadas/inmunología
10.
Immunity ; 43(1): 1-2, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26200004

RESUMEN

The innate immune sensor RIG-I recognizes viral RNA while avoiding unwanted activation by self RNA. In this issue of Immunity, Schuberth-Wagner et al. (2015) show that a histidine residue in the RNA binding pocket of RIG-I sterically excludes the cap1 structure of self RNA, thereby preventing downstream activation.


Asunto(s)
ARN Helicasas DEAD-box/genética , Tolerancia Inmunológica/genética , Procesamiento Postranscripcional del ARN/genética , ARN/genética , Virus de la Fiebre Amarilla/enzimología , Animales , Humanos
11.
J Immunol ; 209(10): 1930-1941, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36426944

RESUMEN

The antiviral state, an initial line of defense against viral infection, is established by a set of IFN-stimulated genes (ISGs) encoding antiviral effector proteins. The effector ISGs are transcriptionally regulated by type I IFNs mainly via activation of IFN-stimulated gene factor 3 (ISGF3). In this study, the regulatory elements of effector ISGs were characterized to determine the (epi)genetic features that enable their robust induction by type I IFNs in multiple cell types. We determined the location of regulatory elements, the DNA motifs, the occupancy of ISGF3 subunits (IRF9, STAT1, and STAT2) and other transcription factors, and the chromatin accessibility of 37 effector ISGs in murine dendritic cells. The IFN-stimulated response element (ISRE) and its tripartite version occurred most frequently in the regulatory elements of effector ISGs than in any other tested ISG subsets. Chromatin accessibility at their promoter regions was similar to most other ISGs but higher than at the promoters of inflammation-related cytokines, which were used as a reference gene set. Most effector ISGs (81.1%) had at least one ISGF3 binding region proximal to the transcription start site (TSS), and only a subset of effector ISGs (24.3%) was associated with three or more ISGF3 binding regions. The IRF9 signals were typically higher, and ISRE motifs were "stronger" (more similar to the canonical sequence) in TSS-proximal versus TSS-distal regulatory regions. Moreover, most TSS-proximal regulatory regions were accessible before stimulation in multiple cell types. Our results indicate that "strong" ISRE motifs and universally accessible promoter regions that permit robust, widespread induction are characteristic features of effector ISGs.


Asunto(s)
Factores de Restricción Antivirales , Cromatina , Animales , Ratones , Cromatina/genética , Motivos de Nucleótidos , Regiones Promotoras Genéticas/genética , Elementos de Respuesta/genética , Interferones/metabolismo
12.
PLoS Genet ; 17(5): e1009578, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34014925

RESUMEN

The Receptor Transporter Protein (RTP) family is present in most, if not all jawed vertebrates. Most of our knowledge of this protein family comes from studies on mammalian RTPs, which are multi-function proteins that regulate cell-surface G-protein coupled receptor levels, influence olfactory system development, regulate immune signaling, and directly inhibit viral infection. However, mammals comprise less than one-tenth of extant vertebrate species, and our knowledge about the expression, function, and evolution of non-mammalian RTPs is limited. Here, we explore the evolutionary history of RTPs in vertebrates. We identify signatures of positive selection in many vertebrate RTP clades and characterize multiple, independent expansions of the RTP family outside of what has been described in mammals. We find a striking expansion of RTPs in the African clawed frog, Xenopus laevis, with 11 RTPs in this species as opposed to 1 to 4 in most other species. RNA sequencing revealed that most X. laevis RTPs are upregulated following immune stimulation. In functional assays, we demonstrate that at least three of these X. laevis RTPs inhibit infection by RNA viruses, suggesting that RTP homologs may serve as antiviral effectors outside of Mammalia.


Asunto(s)
Antivirales , Evolución Molecular , Genómica , Proteínas de Transporte de Membrana/genética , Proteínas de Xenopus/genética , Xenopus laevis/genética , Animales , Antivirales/inmunología , Proteínas de Transporte de Membrana/inmunología , Poli I-C/inmunología , Sintenía , Proteínas de Xenopus/inmunología , Xenopus laevis/inmunología , Xenopus laevis/metabolismo
13.
Proc Natl Acad Sci U S A ; 118(49)2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34873063

RESUMEN

Flaviviruses such as Zika virus and West Nile virus have the potential to cause severe neuropathology if they invade the central nervous system. The type I interferon response is well characterized as contributing to control of flavivirus-induced neuropathogenesis. However, the interferon-stimulated gene (ISG) effectors that confer these neuroprotective effects are less well studied. Here, we used an ISG expression screen to identify Shiftless (SHFL, C19orf66) as a potent inhibitor of diverse positive-stranded RNA viruses, including multiple members of the Flaviviridae (Zika, West Nile, dengue, yellow fever, and hepatitis C viruses). In cultured cells, SHFL functions as a viral RNA-binding protein that inhibits viral replication at a step after primary translation of the incoming genome. The murine ortholog, Shfl, is expressed constitutively in multiple tissues, including the central nervous system. In a mouse model of Zika virus infection, Shfl-/- knockout mice exhibit reduced survival, exacerbated neuropathological outcomes, and increased viral replication in the brain and spinal cord. These studies demonstrate that Shfl is an important antiviral effector that contributes to host protection from Zika virus infection and virus-induced neuropathological disease.


Asunto(s)
Proteínas de Unión al ARN/metabolismo , Infección por el Virus Zika/patología , Virus Zika/metabolismo , Animales , Línea Celular , Efecto Citopatogénico Viral , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/metabolismo , Susceptibilidad a Enfermedades/virología , Flavivirus/genética , Infecciones por Flavivirus/genética , Infecciones por Flavivirus/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fármacos Neuroprotectores/metabolismo , Proteínas de Unión al ARN/genética , Replicación Viral/fisiología , Virus Zika/patogenicidad , Infección por el Virus Zika/genética
14.
J Virol ; 96(19): e0133222, 2022 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-36106874

RESUMEN

Mammalian TRIM7 is an antiviral protein that inhibits multiple human enteroviruses by degrading the viral 2BC protein. Whether TRIM7 is reciprocally targeted by enteroviruses is not known. Here, we report that the 3C protease (3Cpro) from two enteroviruses, coxsackievirus B3 (CVB3) and poliovirus, targets TRIM7 for cleavage. CVB3 3Cpro cleaves TRIM7 at glutamine 24 (Q24), resulting in a truncated TRIM7 that fails to inhibit CVB3 due to dampened E3 ubiquitin ligase activity. TRIM7 Q24 is highly conserved across mammals, except in marsupials, which instead have a naturally occurring histidine (H24) that is not subject to 3Cpro cleavage. Marsupials also express two isoforms of TRIM7, and the two proteins from koalas have distinct antiviral activities. The longer isoform contains an additional exon due to alternate splice site usage. This additional exon contains a unique 3Cpro cleavage site, suggesting that certain enteroviruses may have evolved to target marsupial TRIM7 even if the canonical Q24 is missing. Combined with computational analyses indicating that TRIM7 is rapidly evolving, our data raise the possibility that TRIM7 may be targeted by enterovirus evasion strategies and that evolution of TRIM7 across mammals may have conferred unique antiviral properties. IMPORTANCE Enteroviruses are significant human pathogens that cause viral myocarditis, pancreatitis, and meningitis. Knowing how the host controls these viruses and how the viruses may evade host restriction is important for understanding fundamental concepts in antiviral immunity and for informing potential therapeutic interventions. In this study, we demonstrate that coxsackievirus B3 uses its virally encoded protease to target the host antiviral protein TRIM7 for cleavage, suggesting a potential mechanism of viral immune evasion. We additionally show that TRIM7 has evolved in certain mammalian lineages to express protein variants with distinct antiviral activities and susceptibilities to viral protease-mediated cleavage.


Asunto(s)
Proteasas Virales 3C , Infecciones por Enterovirus , Enterovirus , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Proteasas Virales 3C/metabolismo , Animales , Enterovirus/enzimología , Glutamina , Histidina , Interacciones Huésped-Patógeno , Phascolarctidae/virología , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
15.
PLoS Comput Biol ; 18(4): e1010053, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35468127

RESUMEN

In HIV-1-infected individuals, transmitted/founder (TF) virus contributes to establish new infection and expands during the acute phase of infection, while chronic control (CC) virus emerges during the chronic phase of infection. TF viruses are more resistant to interferon-alpha (IFN-α)-mediated antiviral effects than CC virus, however, its virological relevance in infected individuals remains unclear. Here we perform an experimental-mathematical investigation and reveal that IFN-α strongly inhibits cell-to-cell infection by CC virus but only weakly affects that by TF virus. Surprisingly, IFN-α enhances cell-free infection of HIV-1, particularly that of CC virus, in a virus-cell density-dependent manner. We further demonstrate that LY6E, an IFN-stimulated gene, can contribute to the density-dependent enhancement of cell-free HIV-1 infection. Altogether, our findings suggest that the major difference between TF and CC viruses can be explained by their resistance to IFN-α-mediated inhibition of cell-to-cell infection and their sensitivity to IFN-α-mediated enhancement of cell-free infection.


Asunto(s)
Infecciones por VIH , VIH-1 , Antivirales , Infecciones por VIH/tratamiento farmacológico , Humanos , Interferón-alfa/farmacología
16.
Clin Immunol ; 224: 108662, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33412294

RESUMEN

X-linked severe combined immunodeficiency (X-SCID) is a disorder of adaptive immunity caused by mutations in the IL-2 receptor common gamma chain gene resulting in deficiencies of T and natural killer cells, coupled with severe dysfunction in B cells. X-SCID is lethal without allogeneic stem cell transplant or gene therapy due to opportunistic infections. An infant with X-SCID became infected with SARS-CoV-2 while awaiting transplant. The patient developed severe hepatitis without the respiratory symptoms typical of COVID-19. He was treated with convalescent plasma, and thereafter was confirmed to have SARS-CoV-2 specific antibodies, as detected with a microfluidic antigen array. After resolution of the hepatitis, he received a haploidentical CD34 selected stem cell transplant, without conditioning, from his father who had recovered from COVID-19. SARS CoV-2 was detected via RT-PCR on nasopharyngeal swabs until 61 days post transplantation. He successfully engrafted donor T and NK cells, and continues to do well clinically.


Asunto(s)
COVID-19/complicaciones , COVID-19/terapia , Hepatitis/virología , Inmunodeficiencia Combinada Grave/complicaciones , Humanos , Inmunización Pasiva/métodos , Lactante , Masculino , SARS-CoV-2 , Sueroterapia para COVID-19
17.
J Immunol ; 200(1): 209-217, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29180486

RESUMEN

Bats host a large number of zoonotic viruses, including several viruses that are highly pathogenic to other mammals. The mechanisms underlying this rich viral diversity are unknown, but they may be linked to unique immunological features that allow bats to act as asymptomatic viral reservoirs. Vertebrates respond to viral infection by inducing IFNs, which trigger antiviral defenses through IFN-stimulated gene (ISG) expression. Although the IFN system of several bats is characterized at the genomic level, less is known about bat IFN-mediated transcriptional responses. In this article, we show that IFN signaling in bat cells from the black flying fox (Pteropus alecto) consists of conserved and unique ISG expression profiles. In IFN-stimulated cells, bat ISGs comprise two unique temporal subclusters with similar early induction kinetics but distinct late-phase declines. In contrast, human ISGs lack this decline phase and remained elevated for longer periods. Notably, in unstimulated cells, bat ISGs were expressed more highly than their human counterparts. We also found that the antiviral effector 2-5A-dependent endoribonuclease, which is not an ISG in humans, is highly IFN inducible in black flying fox cells and contributes to cell-intrinsic control of viral infection. These studies reveal distinctive innate immune features that may underlie a unique virus-host relationship in bats.


Asunto(s)
Antivirales/metabolismo , Quirópteros/inmunología , Endorribonucleasas/metabolismo , Factores Reguladores del Interferón/metabolismo , Virosis/inmunología , Animales , Enfermedades Asintomáticas , Línea Celular , Reservorios de Enfermedades , Endorribonucleasas/genética , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Factores Reguladores del Interferón/genética , Interferones/metabolismo , Transducción de Señal
18.
Nature ; 505(7485): 691-5, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24284630

RESUMEN

The type I interferon (IFN) response protects cells from viral infection by inducing hundreds of interferon-stimulated genes (ISGs), some of which encode direct antiviral effectors. Recent screening studies have begun to catalogue ISGs with antiviral activity against several RNA and DNA viruses. However, antiviral ISG specificity across multiple distinct classes of viruses remains largely unexplored. Here we used an ectopic expression assay to screen a library of more than 350 human ISGs for effects on 14 viruses representing 7 families and 11 genera. We show that 47 genes inhibit one or more viruses, and 25 genes enhance virus infectivity. Comparative analysis reveals that the screened ISGs target positive-sense single-stranded RNA viruses more effectively than negative-sense single-stranded RNA viruses. Gene clustering highlights the cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS, also known as MB21D1) as a gene whose expression also broadly inhibits several RNA viruses. In vitro, lentiviral delivery of enzymatically active cGAS triggers a STING-dependent, IRF3-mediated antiviral program that functions independently of canonical IFN/STAT1 signalling. In vivo, genetic ablation of murine cGAS reveals its requirement in the antiviral response to two DNA viruses, and an unappreciated contribution to the innate control of an RNA virus. These studies uncover new paradigms for the preferential specificity of IFN-mediated antiviral pathways spanning several virus families.


Asunto(s)
Inmunidad Innata/genética , Inmunidad Innata/inmunología , Interferones/inmunología , Nucleotidiltransferasas/inmunología , Nucleotidiltransferasas/metabolismo , Virus/inmunología , Animales , Análisis por Conglomerados , Virus ADN/inmunología , Virus ADN/patogenicidad , Citometría de Flujo , Biblioteca de Genes , Factor 3 Regulador del Interferón/inmunología , Factor 3 Regulador del Interferón/metabolismo , Interferones/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Nucleotidiltransferasas/deficiencia , Nucleotidiltransferasas/genética , Virus ARN/inmunología , Virus ARN/patogenicidad , Factor de Transcripción STAT1/metabolismo , Especificidad por Sustrato , Virus/clasificación , Virus/patogenicidad
19.
Nature ; 502(7472): 563-6, 2013 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-24121441

RESUMEN

HIV-1 replication can be inhibited by type I interferon (IFN), and the expression of a number of gene products with anti-HIV-1 activity is induced by type I IFN. However, none of the known antiretroviral proteins can account for the ability of type I IFN to inhibit early, preintegration phases of the HIV-1 replication cycle in human cells. Here, by comparing gene expression profiles in cell lines that differ in their ability to support the inhibitory action of IFN-α at early steps of the HIV-1 replication cycle, we identify myxovirus resistance 2 (MX2) as an interferon-induced inhibitor of HIV-1 infection. Expression of MX2 reduces permissiveness to a variety of lentiviruses, whereas depletion of MX2 using RNA interference reduces the anti-HIV-1 potency of IFN-α. HIV-1 reverse transcription proceeds normally in MX2-expressing cells, but 2-long terminal repeat circular forms of HIV-1 DNA are less abundant, suggesting that MX2 inhibits HIV-1 nuclear import, or destabilizes nuclear HIV-1 DNA. Consistent with this notion, mutations in the HIV-1 capsid protein that are known, or suspected, to alter the nuclear import pathways used by HIV-1 confer resistance to MX2, whereas preventing cell division increases MX2 potency. Overall, these findings indicate that MX2 is an effector of the anti-HIV-1 activity of type-I IFN, and suggest that MX2 inhibits HIV-1 infection by inhibiting capsid-dependent nuclear import of subviral complexes.


Asunto(s)
Infecciones por VIH/prevención & control , VIH-1/fisiología , Interferón-alfa/inmunología , Proteínas de Resistencia a Mixovirus/metabolismo , Transporte Activo de Núcleo Celular , Cápside/metabolismo , División Celular , Línea Celular , Núcleo Celular/metabolismo , Núcleo Celular/virología , Células Cultivadas , Infecciones por VIH/genética , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , VIH-1/inmunología , Humanos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas de Resistencia a Mixovirus/genética , Interferencia de ARN , Transcripción Reversa , Transcriptoma , Replicación Viral
20.
J Infect Dis ; 217(4): 650-655, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29165633

RESUMEN

Polymorphisms at IFNL4 strongly influence spontaneous resolution and interferon therapeutic response in hepatitis C virus (HCV) infection. In chronic HCV, unfavorable alleles are associated with elevated interferon (IFN)-stimulated gene (ISG) expression in the liver, but extrahepatic effects are less well characterized. We used RNA sequencing (RNA-Seq) to examine whether IFNL4 genetic variation (rs368234815) modulates ISG expression in peripheral blood mononuclear cells (PBMC) during chronic HCV infection. ISG expression was elevated in unstimulated PBMC homozygous for the unfavorable ΔG IFNL4 variant; expression following IFN-α stimulation was comparable across genotypes. These findings suggest that lambda interferons may have broader systemic effects during HCV infection.


Asunto(s)
Regulación de la Expresión Génica , Variación Genética , Hepatitis C Crónica/patología , Factores Inmunológicos/biosíntesis , Interleucinas/genética , Células Sanguíneas/inmunología , Perfilación de la Expresión Génica , Humanos , Interferón-alfa/metabolismo , Leucocitos Mononucleares/inmunología , Análisis de Secuencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA