Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Ther Oncolytics ; 28: 321-333, 2023 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-36938543

RESUMEN

Oncolytic viruses (OVs) promote the anti-tumor immune response as their replication, and the subsequent lysis of tumor cells, triggers the activation of immune-sensing pathways. Arming OVs by expressing transgenes with the potential to promote immune cell recruitment and activation is an attractive strategy to enhance OVs' therapeutic benefit. For picornaviruses, a family of OVs with clinical experience, the expression of a transgene is limited by multiple factors: genome physical packaging limits, high rates of recombination, and viral-mediated inhibition of transgene secretion. Here, we evaluated strategies for arming Seneca Valley virus (SVV) with relevant immunomodulatory transgenes. Specificially in the contex of arming SVV, we evaluated transgene maximum size and stabiltity, transgene secretion, and the impact of transgene inclusion on viral fitness. We find that SVV is not capable of expressing secreted payloads and has a transgene packaging capacity of ∼10% of viral genome size. To enable transgene expression, we developed SVV replicons with greater transgene size capacity and secretion capabilities. SVV replicons can be packaged in trans by virus in co-infected cells to express immunomodulatory transgenes in surrounding cells, thus providing a means to enhance the potential of this therapeutic to augment the anti-tumor immune response.

2.
Nat Commun ; 13(1): 5907, 2022 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-36207308

RESUMEN

The therapeutic effectiveness of oncolytic viruses (OVs) delivered intravenously is limited by the development of neutralizing antibody responses against the virus. To circumvent this limitation and to enable repeated systemic administration of OVs, here we develop Synthetic RNA viruses consisting of a viral RNA genome (vRNA) formulated within lipid nanoparticles. For two Synthetic RNA virus drug candidates, Seneca Valley virus (SVV) and Coxsackievirus A21, we demonstrate vRNA delivery and replication, virus assembly, spread and lysis of tumor cells leading to potent anti-tumor efficacy, even in the presence of OV neutralizing antibodies in the bloodstream. Synthetic-SVV replication in tumors promotes immune cell infiltration, remodeling of the tumor microenvironment, and enhances the activity of anti-PD-1 checkpoint inhibitor. In mouse and non-human primates, Synthetic-SVV is well tolerated reaching exposure well above the requirement for anti-tumor activity. Altogether, the Synthetic RNA virus platform provides an approach that enables repeat intravenous administration of viral immunotherapy.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Picornaviridae , Animales , Anticuerpos Neutralizantes , Inmunoterapia , Liposomas , Ratones , Nanopartículas , Neoplasias/terapia , Virus Oncolíticos/genética , ARN Viral/genética , Microambiente Tumoral
3.
Genes Dev ; 35(13-14): 992-1004, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34140354

RESUMEN

Previous work has demonstrated that the epitranscriptomic addition of m6A to viral transcripts can promote the replication and pathogenicity of a wide range of DNA and RNA viruses, including HIV-1, yet the underlying mechanisms responsible for this effect have remained unclear. It is known that m6A function is largely mediated by cellular m6A binding proteins or readers, yet how these regulate viral gene expression in general, and HIV-1 gene expression in particular, has been controversial. Here, we confirm that m6A addition indeed regulates HIV-1 RNA expression and demonstrate that this effect is largely mediated by the nuclear m6A reader YTHDC1 and the cytoplasmic m6A reader YTHDF2. Both YTHDC1 and YTHDF2 bind to multiple distinct and overlapping sites on the HIV-1 RNA genome, with YTHDC1 recruitment serving to regulate the alternative splicing of HIV-1 RNAs. Unexpectedly, while YTHDF2 binding to m6A residues present on cellular mRNAs resulted in their destabilization as previously reported, YTHDF2 binding to m6A sites on HIV-1 transcripts resulted in a marked increase in the stability of these viral RNAs. Thus, YTHDF2 binding can exert diametrically opposite effects on RNA stability, depending on RNA sequence context.


Asunto(s)
VIH-1 , Adenosina/metabolismo , Empalme Alternativo , VIH-1/genética , VIH-1/metabolismo , Empalme del ARN , Estabilidad del ARN/genética , ARN Viral/genética , ARN Viral/metabolismo , Proteínas de Unión al ARN/metabolismo
4.
Cancer Immunol Res ; 9(3): 291-308, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33355229

RESUMEN

ONCR-177 is an engineered recombinant oncolytic herpes simplex virus (HSV) with complementary safety mechanisms, including tissue-specific miRNA attenuation and mutant UL37 to inhibit replication, neuropathic activity, and latency in normal cells. ONCR-177 is armed with five transgenes for IL12, FLT3LG (extracellular domain), CCL4, and antagonists to immune checkpoints PD-1 and CTLA-4. In vitro assays demonstrated that targeted miRNAs could efficiently suppress ONCR-177 replication and transgene expression, as could the HSV-1 standard-of-care therapy acyclovir. Although ONCR-177 was oncolytic across a panel of human cancer cell lines, including in the presence of type I IFN, replication was suppressed in human pluripotent stem cell-derived neurons, cardiomyocytes, and hepatocytes. Dendritic cells activated with ONCR-177 tumor lysates efficiently stimulated tumor antigen-specific CD8+ T-cell responses. In vivo, biodistribution analyses suggested that viral copy number and transgene expression peaked approximately 24 to 72 hours after injection and remained primarily within the injected tumor. Intratumoral administration of ONCR-177 mouse surrogate virus, mONCR-171, was efficacious across a panel of syngeneic bilateral mouse tumor models, resulting in partial or complete tumor regressions that translated into significant survival benefits and to the elicitation of a protective memory response. Antitumor effects correlated with local and distant intratumoral infiltration of several immune effector cell types, consistent with the proposed functions of the transgenes. The addition of systemic anti-PD-1 augmented the efficacy of mONCR-171, particularly for abscopal tumors. Based in part upon these preclinical results, ONCR-177 is being evaluated in patients with metastatic cancer (ONCR-177-101, NCT04348916).


Asunto(s)
Herpesvirus Humano 1/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Animales , Línea Celular Tumoral/trasplante , Terapia Combinada/métodos , Modelos Animales de Enfermedad , Femenino , Herpesvirus Humano 1/genética , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inyecciones Intralesiones , Ratones , MicroARNs/genética , MicroARNs/inmunología , Neoplasias/inmunología , Neoplasias/patología , Virus Oncolíticos/genética , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/metabolismo , Distribución Tisular , Transgenes/genética , Transgenes/inmunología , Proteínas Estructurales Virales/genética , Proteínas Estructurales Virales/inmunología , Replicación Viral/genética
5.
Mol Ther Oncolytics ; 18: 476-490, 2020 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-32953982

RESUMEN

Development of next-generation oncolytic viruses requires the design of vectors that are potently oncolytic, immunogenic in human tumors, and well tolerated in patients. Starting with a joint-region deleted herpes simplex virus 1 (HSV-1) to create large transgene capability, we retained a single copy of the ICP34.5 gene, introduced mutations in UL37 to inhibit retrograde axonal transport, and inserted cell-type-specific microRNA (miRNA) target cassettes in HSV-1 genes essential for replication or neurovirulence. Ten miRNA candidates highly expressed in normal tissues and with low or absent expression in malignancies were selected from a comprehensive profile of 800 miRNAs with an emphasis on protection of the nervous system. Among the genes essential for viral replication identified using a small interfering RNA (siRNA) screen, we selected ICP4, ICP27, and UL8 for miRNA attenuation where a single miRNA is sufficient to potently attenuate viral replication. Additionally, a neuron-specific miRNA target cassette was introduced to control ICP34.5 expression. This vector is resistant to type I interferon compared to ICP34.5-deleted oncolytic HSVs, and in cancer cell lines, the oncolytic activity of the modified vector is equivalent to its parental virus. In vivo, this vector potently inhibits tumor growth while being well tolerated, even at high intravenous doses, compared to parental wild-type HSV-1.

6.
Cell Host Microbe ; 26(2): 217-227.e6, 2019 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-31415754

RESUMEN

How the covalent modification of mRNA ribonucleotides, termed epitranscriptomic modifications, alters mRNA function remains unclear. One issue has been the difficulty of quantifying these modifications. Using purified HIV-1 genomic RNA, we show that this RNA bears more epitranscriptomic modifications than the average cellular mRNA, with 5-methylcytosine (m5C) and 2'O-methyl modifications being particularly prevalent. The methyltransferase NSUN2 serves as the primary writer for m5C on HIV-1 RNAs. NSUN2 inactivation inhibits not only m5C addition to HIV-1 transcripts but also viral replication. This inhibition results from reduced HIV-1 protein, but not mRNA, expression, which in turn correlates with reduced ribosome binding to viral mRNAs. In addition, loss of m5C dysregulates the alternative splicing of viral RNAs. These data identify m5C as a post-transcriptional regulator of both splicing and function of HIV-1 mRNA, thereby affecting directly viral gene expression.


Asunto(s)
5-Metilcitosina/farmacología , Regulación Viral de la Expresión Génica , VIH-1/genética , ARN Viral/metabolismo , Transcriptoma , 5-Metilcitosina/metabolismo , Linfocitos T CD4-Positivos , Regulación Viral de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , Metiltransferasas/farmacología , Empalme del ARN , ARN Mensajero/metabolismo , ARN Viral/efectos de los fármacos , Virión , Replicación Viral/efectos de los fármacos
7.
Disaster Med Public Health Prep ; 13(5-6): 995-1010, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31203830

RESUMEN

A national need is to prepare for and respond to accidental or intentional disasters categorized as chemical, biological, radiological, nuclear, or explosive (CBRNE). These incidents require specific subject-matter expertise, yet have commonalities. We identify 7 core elements comprising CBRNE science that require integration for effective preparedness planning and public health and medical response and recovery. These core elements are (1) basic and clinical sciences, (2) modeling and systems management, (3) planning, (4) response and incident management, (5) recovery and resilience, (6) lessons learned, and (7) continuous improvement. A key feature is the ability of relevant subject matter experts to integrate information into response operations. We propose the CBRNE medical operations science support expert as a professional who (1) understands that CBRNE incidents require an integrated systems approach, (2) understands the key functions and contributions of CBRNE science practitioners, (3) helps direct strategic and tactical CBRNE planning and responses through first-hand experience, and (4) provides advice to senior decision-makers managing response activities. Recognition of both CBRNE science as a distinct competency and the establishment of the CBRNE medical operations science support expert informs the public of the enormous progress made, broadcasts opportunities for new talent, and enhances the sophistication and analytic expertise of senior managers planning for and responding to CBRNE incidents.


Asunto(s)
Derrame de Material Biológico/prevención & control , Liberación de Peligros Químicos/prevención & control , Servicios Médicos de Urgencia/métodos , Sustancias Explosivas/efectos adversos , Liberación de Radiactividad Peligrosa/prevención & control , Planificación en Desastres/organización & administración , Planificación en Desastres/tendencias , Servicios Médicos de Urgencia/tendencias , Humanos
8.
mBio ; 10(3)2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31186331

RESUMEN

While it has been known for several years that viral RNAs are subject to the addition of several distinct covalent modifications to individual nucleotides, collectively referred to as epitranscriptomic modifications, the effect of these editing events on viral gene expression has been controversial. Here, we report the purification of murine leukemia virus (MLV) genomic RNA to homogeneity and show that this viral RNA contains levels of N6-methyladenosine (m6A), 5-methylcytosine (m5C), and 2'O-methylated (Nm) ribonucleotides that are an order of magnitude higher than detected on bulk cellular mRNAs. Mapping of m6A and m5C residues on MLV transcripts identified multiple discrete editing sites and allowed the construction of MLV variants bearing silent mutations that removed a subset of these sites. Analysis of the replication potential of these mutants revealed a modest but significant attenuation in viral replication in 3T3 cells in culture. Consistent with a positive role for m6A and m5C in viral replication, we also demonstrate that overexpression of the key m6A reader protein YTHDF2 enhances MLV replication, while downregulation of the m5C writer NSUN2 inhibits MLV replication.IMPORTANCE The data presented in the present study demonstrate that MLV RNAs bear an exceptionally high level of the epitranscriptomic modifications m6A, m5C, and Nm, suggesting that these each facilitate some aspect of the viral replication cycle. Consistent with this hypothesis, we demonstrate that mutational removal of a subset of these m6A or m5C modifications from MLV transcripts inhibits MLV replication in cis, and a similar result was also observed upon manipulation of the level of expression of key cellular epitranscriptomic cofactors in trans Together, these results argue that the addition of several different epitranscriptomic modifications to viral transcripts stimulates viral gene expression and suggest that MLV has therefore evolved to maximize the level of these modifications that are added to viral RNAs.


Asunto(s)
Adenosina/química , Citosina/química , Metilación de ADN , Virus de la Leucemia Murina/genética , Replicación Viral , Expresión Génica , Genoma Viral , Virus de la Leucemia Murina/fisiología , Metiltransferasas/metabolismo , ARN Mensajero , ARN Viral/genética
9.
Future Virol ; 13(7): 475-482, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30245733

RESUMEN

AIM: The goal of this study was to determine if a single AAV vector, encoding Cas9 and guide RNAs specific for the HPV16 E6 and E7 genes, could inhibit the growth of an HPV16-induced tumor in vivo. MATERIALS & METHODS: We grew HPV16+, patient-derived anal cancer explants in immunodeficient mice and then challenged these by injection of AAV-based vectors encoding Cas9 and control or HPV16-specific guide RNAs. RESULTS & CONCLUSION: We observed a significant and selective reduction in tumor growth when the HPV16 E6 and E7 genes were targeted using Cas9. These studies provide proof of principle for the hypothesis that CRISPR/Cas has the potential to be used to selectively treat HPV-induced tumors in humans.

10.
RNA ; 24(9): 1172-1182, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29903832

RESUMEN

While the issue of whether RNA interference (RNAi) ever forms part of the antiviral innate immune response in mammalian somatic cells remains controversial, there is considerable evidence demonstrating that few, if any, viral small interfering RNAs (siRNAs) are produced in infected cells. Moreover, inhibition of RNAi by mutational inactivation of key RNAi factors, such as Dicer or Argonaute 2, fails to enhance virus replication. One potential explanation for this lack of inhibitory effect is that mammalian viruses encode viral suppressors of RNAi (VSRs) that are so effective that viral siRNAs are not produced in infected cells. Indeed, a number of mammalian VSRs have been described, of which the most prominent is the influenza A virus (IAV) NS1 protein, which has not only been reported to inhibit RNAi in plants and insects but also to prevent the production of viral siRNAs in IAV-infected human cells. Here, we confirm that an IAV mutant lacking NS1 indeed differs from wild-type IAV in that it induces the production of readily detectable levels of Dicer-dependent viral siRNAs in infected human cells. However, we also demonstrate that these siRNAs have little if any inhibitory effect on IAV gene expression. This is likely due, at least in part, to their inefficient loading into RNA-induced silencing complexes.


Asunto(s)
ARN Helicasas DEAD-box/genética , Virus de la Influenza A/fisiología , Interferencia de ARN , Ribonucleasa III/genética , Proteínas no Estructurales Virales/genética , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Virus de la Influenza A/genética , Mutación , ARN Viral/genética , Análisis de Secuencia de ARN , Replicación Viral
12.
Cell Host Microbe ; 22(3): 377-386.e5, 2017 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-28910636

RESUMEN

Many viral RNAs are modified by methylation of the N6 position of adenosine (m6A). m6A is thought to regulate RNA splicing, stability, translation, and secondary structure. Influenza A virus (IAV) expresses m6A-modified RNAs, but the effects of m6A on this segmented RNA virus remain unclear. We demonstrate that global inhibition of m6A addition inhibits IAV gene expression and replication. In contrast, overexpression of the cellular m6A "reader" protein YTHDF2 increases IAV gene expression and replication. To address whether m6A residues modulate IAV RNA function in cis, we mapped m6A residues on the IAV plus (mRNA) and minus (vRNA) strands and used synonymous mutations to ablate m6A on both strands of the hemagglutinin (HA) segment. These mutations inhibited HA mRNA and protein expression while leaving other IAV mRNAs and proteins unaffected, and they also resulted in reduced IAV pathogenicity in mice. Thus, m6A residues in IAV transcripts enhance viral gene expression.


Asunto(s)
Virus de la Influenza A/fisiología , Gripe Humana/virología , ARN Viral/genética , Replicación Viral , Animales , Metilación de ADN , Epigénesis Genética , Femenino , Interacciones Huésped-Patógeno , Humanos , Virus de la Influenza A/genética , Gripe Humana/genética , Gripe Humana/metabolismo , Metilación , Ratones Endogámicos C57BL , ARN Viral/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transcriptoma
13.
Cell Rep ; 20(7): 1503-1512, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28813663

RESUMEN

Influenza A virus (IAV) is a pathogen that poses significant risks to human health. It is therefore critical to develop strategies to prevent influenza disease. Many loss-of-function screens have been performed to identify the host proteins required for viral infection. However, there has been no systematic screen to identify the host factors that, when overexpressed, are sufficient to prevent infection. In this study, we used CRISPR/dCas9 activation technology to perform a genome-wide overexpression screen to identify IAV restriction factors. The major hit from our screen, B4GALNT2, showed inhibitory activity against influenza viruses with an α2,3-linked sialic acid receptor preference. B4GALNT2 overexpression prevented the infection of every avian influenza virus strain tested, including the H5, H9, and H7 subtypes, which have previously caused disease in humans. Thus, we have used CRISPR/dCas9 activation technology to identify a factor that can abolish infection by avian influenza viruses.


Asunto(s)
Sistemas CRISPR-Cas , Interacciones Huésped-Parásitos/inmunología , Virus de la Influenza A/inmunología , N-Acetilgalactosaminiltransferasas/inmunología , Receptores Virales/inmunología , Ácidos Siálicos/inmunología , Células A549 , Animales , Secuencia de Carbohidratos , Perros , Expresión Génica , Genes Reporteros , Ingeniería Genética , Genoma Humano , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Virus de la Influenza A/clasificación , Virus de la Influenza A/genética , Luciferasas/genética , Luciferasas/metabolismo , Células de Riñón Canino Madin Darby , N-Acetilgalactosaminiltransferasas/química , N-Acetilgalactosaminiltransferasas/genética , Polisacáridos/química , Polisacáridos/inmunología , Polisacáridos/metabolismo , Receptores Virales/química , Receptores Virales/genética , Ácidos Siálicos/química , Ácidos Siálicos/metabolismo
14.
RNA Biol ; 14(11): 1570-1579, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-28594311

RESUMEN

While lentiviral expression vectors are widely used in many facets of molecular biology, due to their ability to stably express heterologous genes in both dividing and non-dividing cells, they suffer from the disadvantage that introns inserted into the vector genome are generally rapidly lost by splicing in packaging cell lines. The presence of an intron, if achievable, has the potential to facilitate the expression of transgene cDNAs, as splicing has been extensively shown to facilitate mRNA biogenesis and function. Moreover, if a stable intron could be introduced into a lentiviral vector, this could greatly facilitate the expression of microRNAs (miRNAs), and especially miRNA clusters, as the introduction of pri-miRNA stems into the exonic region of a lentiviral vector can strongly reduce both vector titer and the expression of any miRNA-linked indicator gene due to cleavage of the vector RNA genome by cellular Drosha. Here, we describe a novel lentiviral vector design in which transgenes and/or miRNAs are expressed using an antisense-orientated, inducible promoter driving an expression cassette bearing a functional intron. We demonstrate that this lentiviral vector, called pTREX, is able to express higher levels of both transgenes and pri-miRNA clusters when compared with a closely similar conventional lentiviral vector.


Asunto(s)
Ingeniería Genética/métodos , Intrones , Lentivirus/genética , MicroARNs/genética , Empalme del ARN , Línea Celular , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Exones , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Humanos , Insulina/genética , Insulina/metabolismo , Lentivirus/metabolismo , MicroARNs/metabolismo , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Plásmidos/química , Plásmidos/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Regiones Promotoras Genéticas , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Antígenos Thy-1/genética , Antígenos Thy-1/metabolismo , Transgenes , eIF-2 Quinasa/deficiencia , eIF-2 Quinasa/genética
15.
J Virol ; 91(9)2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28250115

RESUMEN

Although it has been known for over 40 years that eukaryotic mRNAs bear internal base modifications, it is only in the last 5 years that the importance of these modifications has begun to come into focus. The most common mRNA modification, the addition of a methyl group to the N6 position of adenosine (m6A), has been shown to affect splicing, translation, and stability, and m6A is also essential for embryonic development in organisms ranging from plants to mice. While all viral transcripts examined so far have been found to be extensively m6A modified, the role, if any, of m6A in regulating viral gene expression and replication was previously unknown. However, recent data generated using HIV-1 as a model system strongly suggest that sites of m6A addition not only are evolutionarily conserved but also enhance virus replication. It is therefore likely that the field of viral epitranscriptomics, which can be defined as the study of functionally relevant posttranscriptional modifications of viral RNA transcripts that do not change the nucleotide sequence of that RNA, is poised for a major expansion in scientific interest and may well fundamentally change our understanding of how viral replication is regulated.


Asunto(s)
Adenosina/análogos & derivados , Virus ADN/genética , Procesamiento Postranscripcional del ARN/genética , Virus ARN/genética , ARN Mensajero/genética , ARN Viral/genética , Adenosina/genética , VIH-1/genética , Humanos , Metilación , Metiltransferasas/metabolismo
16.
Cell ; 169(1): 72-84.e13, 2017 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-28340352

RESUMEN

Multiple sclerosis (MS) is an autoimmune disorder where T cells attack neurons in the central nervous system (CNS) leading to demyelination and neurological deficits. A driver of increased MS risk is the soluble form of the interleukin-7 receptor alpha chain gene (sIL7R) produced by alternative splicing of IL7R exon 6. Here, we identified the RNA helicase DDX39B as a potent activator of this exon and consequently a repressor of sIL7R, and we found strong genetic association of DDX39B with MS risk. Indeed, we showed that a genetic variant in the 5' UTR of DDX39B reduces translation of DDX39B mRNAs and increases MS risk. Importantly, this DDX39B variant showed strong genetic and functional epistasis with allelic variants in IL7R exon 6. This study establishes the occurrence of biological epistasis in humans and provides mechanistic insight into the regulation of IL7R exon 6 splicing and its impact on MS risk.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Epistasis Genética , Subunidad alfa del Receptor de Interleucina-7/genética , Empalme del ARN , ARN Helicasas DEAD-box/genética , Exones , Células HeLa , Humanos , Esclerosis Múltiple/genética , Biosíntesis de Proteínas , ARN Interferente Pequeño/metabolismo , Linfocitos T/inmunología
17.
mBio ; 8(1)2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28096489

RESUMEN

Analysis of the incorporation of cellular microRNAs (miRNAs) into highly purified HIV-1 virions revealed that this largely, but not entirely, mirrored the level of miRNA expression in the producer CD4+ T cells. Specifically, of the 58 cellular miRNAs detected at significant levels in the producer cells, only 5 were found in virions at a level 2- to 4-fold higher than that predicted on the basis of random cytoplasmic sampling. Of note, these included two miRNAs, miR-155 and miR-92a, that were reported previously to at least weakly bind HIV-1 transcripts. To test whether miRNA binding to the HIV-1 genome can induce virion incorporation, artificial miRNA target sites were introduced into the viral genome and a 10- to 40-fold increase in the packaging of the cognate miRNAs into virions was then observed, leading to the recruitment of up to 1.6 miRNA copies per virion. Importantly, this high level of incorporation significantly inhibited HIV-1 virion infectivity. These results suggest that target sites for cellular miRNAs can inhibit RNA virus replication at two distinct steps, i.e., during infection and during viral gene expression, thus explaining why a range of different RNA viruses appear to have evolved to avoid cellular miRNA binding to their genome. IMPORTANCE: The genomes of RNA viruses have the potential to interact with cellular miRNAs, which could lead to their incorporation into virions, with unknown effects on virion function. Here, it is demonstrated that wild-type HIV-1 virions essentially randomly incorporate low levels of the miRNAs expressed by infected cells. However, the specific incorporation of high levels of individual cellular miRNAs can be induced by insertion of cognate target sites into the viral genome. Of note, this results in a modest but significant inhibition of virion infectivity. These data imply that cellular miRNAs have the potential to inhibit viral replication by interfering with not only viral mRNA function but also virion infectivity.


Asunto(s)
Antivirales/análisis , VIH-1/crecimiento & desarrollo , VIH-1/fisiología , MicroARNs/análisis , Virión/química , Ensamble de Virus , Línea Celular , Humanos
18.
Annu Rev Med ; 68: 401-411, 2017 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-27576009

RESUMEN

The emergence of the CRISPR/Cas system of antiviral adaptive immunity in bacteria as a facile system for gene editing in mammalian cells may well lead to gene editing becoming a novel treatment for a range of human diseases, especially those caused by deleterious germline mutations. Another potential target for gene editing are DNA viruses that cause chronic pathogenic diseases that cannot be cured by using currently available drugs. We review the current state of this field and discuss the potential advantages and problems with using a gene editing approach as a treatment for diseases caused by DNA viruses.


Asunto(s)
Sistemas CRISPR-Cas , Infecciones por Virus ADN/terapia , Virus ADN/genética , Edición Génica , Infecciones por VIH/terapia , VIH-1/genética , Terapia Genética/métodos , Vectores Genéticos , Virus de la Hepatitis B/genética , Hepatitis B Crónica/terapia , Herpes Genital/terapia , Herpes Simple/terapia , Herpesvirus Humano 1/genética , Herpesvirus Humano 2/genética , Humanos , Papillomaviridae/genética , Infecciones por Papillomavirus/terapia
19.
RNA ; 23(2): 153-160, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27837013

RESUMEN

While mammalian somatic cells are incapable of mounting an effective RNA interference (RNAi) response to viral infections, plants and invertebrates are able to generate high levels of viral short interfering RNAs (siRNAs) that can control many infections. In Drosophila, the RNAi response is mediated by the Dicer 2 enzyme (dDcr2) acting in concert with two cofactors called Loqs-PD and R2D2. To examine whether a functional RNAi response could be mounted in human somatic cells, we expressed dDcr2, in the presence or absence of Loqs-PD and/or R2D2, in a previously described human cell line, NoDice/ΔPKR, that lacks functional forms of human Dicer (hDcr) and PKR. We observed significant production of ∼21-nt long siRNAs, derived from a cotransfected double stranded RNA (dsRNA) expression vector, that were loaded into the human RNA-induced silencing complex (RISC) and were able to significantly reduce the expression of a cognate indicator gene. Surprisingly, dDcr2 was able to produce siRNAs even in the absence of Loqs-PD, which is thought to be required for dsRNA cleavage by dDcr2. This result may be explained by our finding that dDcr2 is able to bind the human Loqs-PD homolog TRBP when expressed in human cells in the absence of Loqs-PD. We conclude that it is possible to at least partially rescue the ability of mammalian somatic cells to express functional siRNAs using gene products of invertebrate origin.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , ARN Helicasas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/genética , Ribonucleasa III/genética , Animales , Ingeniería Celular , Línea Celular , ARN Helicasas DEAD-box/deficiencia , ARN Helicasas DEAD-box/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Prueba de Complementación Genética , Humanos , Coactivadores de Receptor Nuclear/genética , Coactivadores de Receptor Nuclear/metabolismo , Unión Proteica , ARN Helicasas/metabolismo , ARN Interferente Pequeño/biosíntesis , Proteínas de Unión al ARN/metabolismo , Complejo Silenciador Inducido por ARN/biosíntesis , Complejo Silenciador Inducido por ARN/genética , Ribonucleasa III/deficiencia , Ribonucleasa III/metabolismo , Transgenes , eIF-2 Quinasa/deficiencia , eIF-2 Quinasa/genética
20.
Cell Host Microbe ; 20(5): 654-665, 2016 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-27773535

RESUMEN

The RNA modification N6-methyladenosine (m6A) post-transcriptionally regulates RNA function. The cellular machinery that controls m6A includes methyltransferases and demethylases that add or remove this modification, as well as m6A-binding YTHDF proteins that promote the translation or degradation of m6A-modified mRNA. We demonstrate that m6A modulates infection by hepatitis C virus (HCV). Depletion of m6A methyltransferases or an m6A demethylase, respectively, increases or decreases infectious HCV particle production. During HCV infection, YTHDF proteins relocalize to lipid droplets, sites of viral assembly, and their depletion increases infectious viral particles. We further mapped m6A sites across the HCV genome and determined that inactivating m6A in one viral genomic region increases viral titer without affecting RNA replication. Additional mapping of m6A on the RNA genomes of other Flaviviridae, including dengue, Zika, yellow fever, and West Nile virus, identifies conserved regions modified by m6A. Altogether, this work identifies m6A as a conserved regulatory mark across Flaviviridae genomes.


Asunto(s)
Adenosina/análogos & derivados , Flaviviridae/genética , Flaviviridae/fisiología , Regulación Viral de la Expresión Génica , Interacciones Huésped-Patógeno , ARN Viral/metabolismo , Replicación Viral , Adenosina/metabolismo , Metiltransferasas/metabolismo , Oxidorreductasas N-Desmetilantes/metabolismo , Carga Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...