Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Nucleic Acids Res ; 49(11): 6082-6099, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34057477

RESUMEN

Oligonucleotide-based therapeutics have the capacity to engage with nucleic acid immune sensors to activate or block their response, but a detailed understanding of these immunomodulatory effects is currently lacking. We recently showed that 2'-O-methyl (2'OMe) gapmer antisense oligonucleotides (ASOs) exhibited sequence-dependent inhibition of sensing by the RNA sensor Toll-Like Receptor (TLR) 7. Here we discovered that 2'OMe ASOs can also display sequence-dependent inhibitory effects on two major sensors of DNA, namely cyclic GMP-AMP synthase (cGAS) and TLR9. Through a screen of 80 2'OMe ASOs and sequence mutants, we characterized key features within the 20-mer ASOs regulating cGAS and TLR9 inhibition, and identified a highly potent cGAS inhibitor. Importantly, we show that the features of ASOs inhibiting TLR9 differ from those inhibiting cGAS, with only a few sequences inhibiting both pathways. Together with our previous studies, our work reveals a complex pattern of immunomodulation where 95% of the ASOs tested inhibited at least one of TLR7, TLR9 or cGAS by ≥30%, which may confound interpretation of their in vivo functions. Our studies constitute the broadest analysis of the immunomodulatory effect of 2'OMe ASOs on nucleic acid sensing to date and will support refinement of their therapeutic development.


Asunto(s)
Nucleotidiltransferasas/antagonistas & inhibidores , Oligonucleótidos Antisentido/química , Receptor Toll-Like 9/antagonistas & inhibidores , Adulto , Animales , Secuencia de Bases , Células Cultivadas , ADN , Humanos , Ratones , Transducción de Señal , Receptor Toll-Like 3/antagonistas & inhibidores , Receptor Toll-Like 7/antagonistas & inhibidores
2.
Nucleic Acids Res ; 48(13): 7052-7065, 2020 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-32544249

RESUMEN

Oligonucleotide-based therapeutics have become a reality, and are set to transform management of many diseases. Nevertheless, the modulatory activities of these molecules on immune responses remain incompletely defined. Here, we show that gene targeting 2'-O-methyl (2'OMe) gapmer antisense oligonucleotides (ASOs) can have opposing activities on Toll-Like Receptors 7 and 8 (TLR7/8), leading to divergent suppression of TLR7 and activation of TLR8, in a sequence-dependent manner. Surprisingly, TLR8 potentiation by the gapmer ASOs was blunted by locked nucleic acid (LNA) and 2'-methoxyethyl (2'MOE) modifications. Through a screen of 192 2'OMe ASOs and sequence mutants, we characterized the structural and sequence determinants of these activities. Importantly, we identified core motifs preventing the immunosuppressive activities of 2'OMe ASOs on TLR7. Based on these observations, we designed oligonucleotides strongly potentiating TLR8 sensing of Resiquimod, which preserve TLR7 function, and promote strong activation of phagocytes and immune cells. We also provide proof-of-principle data that gene-targeting ASOs can be selected to synergize with TLR8 agonists currently under investigation as immunotherapies, and show that rational ASO selection can be used to prevent unintended immune suppression of TLR7. Taken together, our work characterizes the immumodulatory effects of ASOs to advance their therapeutic development.


Asunto(s)
Oligodesoxirribonucleótidos Antisentido/farmacología , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/metabolismo , Células Cultivadas , Humanos , Imidazoles/metabolismo , Leucocitos Mononucleares , Oligonucleótidos/metabolismo , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas
3.
Nucleic Acids Res ; 44(2): 863-77, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26578588

RESUMEN

Thousands of long non-coding RNAs (lncRNAs) have been identified in mammalian cells. Some have important functions and their dysregulation can contribute to a variety of disease states. However, most lncRNAs have not been functionally characterized. Complicating their study, lncRNAs have widely varying subcellular distributions: some reside predominantly in the nucleus, the cytoplasm or in both compartments. One method to query function is to suppress expression and examine the resulting phenotype. Methods to suppress expression of mRNAs include antisense oligonucleotides (ASOs) and RNA interference (RNAi). Antisense and RNAi-based gene-knockdown methods vary in efficacy between different cellular compartments. It is not known if this affects their ability to suppress lncRNAs. To address whether localization of the lncRNA influences susceptibility to degradation by either ASOs or RNAi, nuclear lncRNAs (MALAT1 and NEAT1), cytoplasmic lncRNAs (DANCR and OIP5-AS1) and dual-localized lncRNAs (TUG1, CasC7 and HOTAIR) were compared for knockdown efficiency. We found that nuclear lncRNAs were more effectively suppressed using ASOs, cytoplasmic lncRNAs were more effectively suppressed using RNAi and dual-localized lncRNAs were suppressed using both methods. A mixed-modality approach combining ASOs and RNAi reagents improved knockdown efficacy, particularly for those lncRNAs that localize to both nuclear and cytoplasmic compartments.


Asunto(s)
Técnicas de Silenciamiento del Gen/métodos , Oligonucleótidos Antisentido/genética , Interferencia de ARN , ARN Largo no Codificante/metabolismo , Núcleo Celular/genética , Citoplasma/genética , Células HCT116 , Células HeLa , Humanos , ARN Largo no Codificante/genética
4.
Nucleic Acids Res ; 43(2): 1177-88, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25539920

RESUMEN

Anti-microRNA (miRNA) oligonucleotides (AMOs) with 2'-O-Methyl (2'OMe) residues are commonly used to study miRNA function and can achieve high potency, with low cytotoxicity. Not withstanding this, we demonstrate the sequence-dependent capacity of 2'OMe AMOs to inhibit Toll-like receptor (TLR) 7 and 8 sensing of immunostimulatory RNA, independent of their miRNA-targeting function. Through a screen of 29 AMOs targeting common miRNAs, we found a subset of sequences highly inhibitory to TLR7 sensing in mouse macrophages. Interspecies conservation of this inhibitory activity was confirmed on TLR7/8 activity in human peripheral blood mononuclear cells. Significantly, we identified a core motif governing the inhibitory activity of these AMOs, which is present in more than 50 AMOs targeted to human miRNAs in miRBaseV20. DNA/locked nucleic acids (LNA) AMOs synthesized with a phosphorothioate backbone also inhibited TLR7 sensing in a sequence-dependent manner, demonstrating that the off-target effects of AMOs are not restricted to 2'OMe modification. Taken together, our work establishes the potential for off-target effects of AMOs on TLR7/8 function, which should be taken into account in their therapeutic development and in vivo application.


Asunto(s)
MicroARNs/antagonistas & inhibidores , Oligonucleótidos/química , Receptor Toll-Like 7/antagonistas & inhibidores , Receptor Toll-Like 8/antagonistas & inhibidores , Adyuvantes Inmunológicos/farmacología , Animales , Secuencia de Bases , Células HEK293 , Humanos , Ratones , Motivos de Nucleótidos , ARN/farmacología
5.
EMBO J ; 30(5): 835-45, 2011 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-21285947

RESUMEN

MicroRNAs (miRNAs) were shown to be important for pancreas development, yet their roles in differentiated ß-cells remain unclear. Here, we show that miRNA inactivation in ß-cells of adult mice results in a striking diabetic phenotype. While islet architecture is intact and differentiation markers are maintained, Dicer1-deficient ß-cells show a dramatic decrease in insulin content and insulin mRNA. As a consequence of the change in insulin content, the animals become diabetic. We provide evidence for involvement of a set of miRNAs in regulating insulin synthesis. The specific knockdown of miR-24, miR-26, miR-182 or miR-148 in cultured ß-cells or in isolated primary islets downregulates insulin promoter activity and insulin mRNA levels. Further, miRNA-dependent regulation of insulin expression is associated with upregulation of transcriptional repressors, including Bhlhe22 and Sox6. Thus, miRNAs in the adult pancreas act in a new network that reinforces insulin expression by reducing the expression of insulin transcriptional repressors.


Asunto(s)
ARN Helicasas DEAD-box/fisiología , Endorribonucleasas/fisiología , Células Secretoras de Insulina/metabolismo , Insulina/genética , Insulina/metabolismo , MicroARNs/fisiología , Proteínas Represoras/metabolismo , Transcripción Genética , Animales , Western Blotting , Diferenciación Celular , Células Cultivadas , Regulación hacia Abajo , Intolerancia a la Glucosa , Humanos , Técnicas para Inmunoenzimas , Células Secretoras de Insulina/citología , Integrasas/metabolismo , Luciferasas/metabolismo , Ratones , Ratones Noqueados , MicroARNs/antagonistas & inhibidores , ARN Mensajero/genética , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleasa III
6.
Development ; 139(16): 3021-31, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22764048

RESUMEN

Genome-encoded microRNAs (miRNAs) provide a post-transcriptional regulatory layer that is important for pancreas development. However, how specific miRNAs are intertwined into the transcriptional network, which controls endocrine differentiation, is not well understood. Here, we show that microRNA-7 (miR-7) is specifically expressed in endocrine precursors and in mature endocrine cells. We further demonstrate that Pax6 is an important target of miR-7. miR-7 overexpression in developing pancreas explants or in transgenic mice led to Pax6 downregulation and inhibition of α- and ß-cell differentiation, resembling the molecular changes caused by haploinsufficient expression of Pax6. Accordingly, miR-7 knockdown resulted in Pax6 upregulation and promoted α- and ß-cell differentiation. Furthermore, Pax6 downregulation reversed the effect of miR-7 knockdown on insulin promoter activity. These data suggest a novel miR-7-based circuit that ensures precise control of endocrine cell differentiation.


Asunto(s)
Islotes Pancreáticos/embriología , Islotes Pancreáticos/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Páncreas/embriología , Páncreas/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proteínas del Ojo/antagonistas & inhibidores , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Haploinsuficiencia , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Insulina/genética , Islotes Pancreáticos/citología , Ratones , Ratones Endogámicos ICR , Ratones Transgénicos , MicroARNs/antagonistas & inhibidores , Modelos Biológicos , Técnicas de Cultivo de Órganos , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/antagonistas & inhibidores , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Páncreas/citología , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Regulación hacia Arriba
7.
Proc Natl Acad Sci U S A ; 109(33): 13362-7, 2012 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-22853952

RESUMEN

Production of functional proteins requires multiple steps, including gene transcription and posttranslational processing. MicroRNAs (miRNAs) can regulate individual stages of these processes. Despite the importance of the cystic fibrosis transmembrane conductance regulator (CFTR) channel for epithelial anion transport, how its expression is regulated remains uncertain. We discovered that miRNA-138 regulates CFTR expression through its interactions with the transcriptional regulatory protein SIN3A. Treating airway epithelia with an miR-138 mimic increased CFTR mRNA and also enhanced CFTR abundance and transepithelial Cl(-) permeability independent of elevated mRNA levels. An miR-138 anti-miR had the opposite effects. Importantly, miR-138 altered the expression of many genes encoding proteins that associate with CFTR and may influence its biosynthesis. The most common CFTR mutation, ΔF508, causes protein misfolding, protein degradation, and cystic fibrosis. Remarkably, manipulating the miR-138 regulatory network also improved biosynthesis of CFTR-ΔF508 and restored Cl(-) transport to cystic fibrosis airway epithelia. This miRNA-regulated network directs gene expression from the chromosome to the cell membrane, indicating that an individual miRNA can control a cellular process more broadly than recognized previously. This discovery also provides therapeutic avenues for restoring CFTR function to cells affected by the most common cystic fibrosis mutation.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/biosíntesis , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulación de la Expresión Génica , Redes Reguladoras de Genes/genética , MicroARNs/metabolismo , Transporte Biológico , Cloruros/metabolismo , Epitelio/metabolismo , Epitelio/patología , Perfilación de la Expresión Génica , Células HeLa , Humanos , Pulmón/metabolismo , Pulmón/patología , MicroARNs/genética , Procesamiento Proteico-Postraduccional , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Complejo Correpresor Histona Desacetilasa y Sin3
8.
Am J Respir Cell Mol Biol ; 49(4): 544-51, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23646886

RESUMEN

MicroRNAs (miRNAs) are increasingly recognized as important posttranscriptional regulators of gene expression, and changes in their actions can contribute to disease states. Little is understood regarding miRNA functions in the airway epithelium under normal or diseased conditions. We profiled miRNA expression in well-differentiated primary cultures of human cystic fibrosis (CF) and non-CF airway epithelia, and discovered that miR-509-3p and miR-494 concentrations were increased in CF epithelia. Human non-CF airway epithelia, transfected with the mimics of miR-509-3p or miR-494, showed decreased cystic fibrosis transmembrane conductance regulator (CFTR) expression, whereas their respective anti-miRs exerted the opposite effect. Interestingly, the two miRNAs acted cooperatively in regulating CFTR expression. Upon infecting non-CF airway epithelial cells with Staphylococcus aureus, or upon stimulating them with the proinflammatory cytokines TNF-α or IL-1ß, we observed an increased expression of both miRNAs and a concurrent decrease in CFTR expression and function, suggesting that inflammatory mediators may regulate these miRNAs. Transfecting epithelia with anti-miRs for miR-509-3p and miR-494, or inhibiting NF-κB signaling before stimulating cells with TNFα or IL-1ß, suppressed these responses, suggesting that the expression of both miRNAs was responsive to NF-κB signaling. Thus, miR-509-3p and miR-494 are dynamic regulators of CFTR abundance and function in normal, non-CF airway epithelia.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/metabolismo , MicroARNs/metabolismo , Procesamiento Postranscripcional del ARN , Línea Celular , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Células Epiteliales/metabolismo , Expresión Génica , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , MicroARNs/genética , FN-kappa B/genética , FN-kappa B/metabolismo , Mucosa Respiratoria/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
9.
Neurobiol Dis ; 56: 6-13, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23583610

RESUMEN

Spinocerebellar Ataxia Type 1 (SCA1) is an autosomal dominant late onset neurodegenerative disease caused by an expanded polyglutamine tract in ataxin-1. Here, we compared the protective effects of overexpressing ataxin-1-like using recombinant AAVs, or reducing expression of mutant ataxin-1 using virally delivered RNA interference (RNAi), in a transgenic mouse model of SCA1. For the latter, we used an artificial microRNA (miR) design that optimizes potency, efficacy and safety to suppress ataxin-1 expression (miS1). Delivery of either ataxin-1-like or miS1 viral vectors to SCA1 mice cerebella resulted in widespread cerebellar Purkinje cell transduction and improved behavioral and histological phenotypes. Our data indicate the utility of either approach as a possible therapy for SCA1 patients.


Asunto(s)
Proteínas del Tejido Nervioso/biosíntesis , Proteínas Nucleares/biosíntesis , Interferencia de ARN/fisiología , Ataxias Espinocerebelosas/terapia , Animales , Ataxina-1 , Ataxinas , Conducta Animal/fisiología , Western Blotting , Encéfalo/patología , Dependovirus/genética , Marcha/fisiología , Vectores Genéticos , Células HEK293 , Humanos , Inmunohistoquímica , Inmunoprecipitación , Hibridación in Situ , Locomoción/fisiología , Ratones , Ratones Transgénicos , MicroARNs/biosíntesis , MicroARNs/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Plásmidos , Equilibrio Postural/fisiología , ARN Interferente Pequeño/uso terapéutico , Reacción en Cadena en Tiempo Real de la Polimerasa , Ataxias Espinocerebelosas/patología , Ataxias Espinocerebelosas/psicología
10.
Pharm Res ; 27(9): 1788-99, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20424893

RESUMEN

PURPOSE: Cataloguing endogenous miRNA targets by inhibiting miRNA function is fundamental to understanding the biological importance of each miRNA in gene regulatory pathways. Methods to down-regulate miRNA activity may help treat diseases where over-expression of miRNAs relates to the underlying pathophysiology. This study objectively evaluates the in vitro potency of different anti-miRNA oligonucleotides (AMOs) using various design and modification strategies described in the literature as well as some novel modification strategies. METHODS: MiR21 and miR16 AMOs, containing chemical modifications such as 2'-O-methyl RNA, locked nucleic acid and 2'-Fluoro bases with or without phosphorothioate linkages, were directly compared by transfection into HeLa cells using a dual-luciferase reporter assay to quantify miRNA inhibition. RESULTS: Potency for the various AMOs ranged from inactive at high dose (50 nM) to strongly inhibitory at both high and low dose (1 nM). Including phosphorothioate linkages improved nuclease stability and generally increased functional potency. CONCLUSIONS: Incorporating high binding affinity modifications, such as LNA and 2'F bases, increases AMO potency while maintaining specificity; nevertheless, use of low dose is preferred when using high potency reagents to minimize the potential for cross reactivity. 2'OMe/LNA chimeras with PS modifications were the most potent constructs tested for miRNA inhibition in vitro.


Asunto(s)
MicroARNs/antagonistas & inhibidores , Oligonucleótidos Antisentido/farmacología , Animales , Secuencia de Bases , Sitios de Unión , Técnicas de Cultivo de Célula , Cromatografía Líquida de Alta Presión , Estabilidad de Enzimas , Células HeLa , Humanos , Luciferasas de Luciérnaga/genética , Luciferasas de Renilla/genética , Masculino , Ratones , MicroARNs/genética , Datos de Secuencia Molecular , Oligonucleótidos Antisentido/química , Oligonucleótidos Antisentido/genética , Plásmidos , Ribonucleasas/metabolismo , Espectrometría de Masa por Ionización de Electrospray , Transfección
11.
Methods Mol Biol ; 2176: 121-140, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32865787

RESUMEN

Long noncoding RNAs (lncRNAs) are a recently discovered class of RNA that have diverse intracellular regulatory and structural roles. Because of their wide assortment of functions, lncRNAs can have varied distributions in the nucleus and/or cytoplasm of a cell. However, even though tens of thousands of human lncRNAs have been identified, currently less than 3% have empirically validated functions. RNA knockdown is now a relatively commonplace laboratory technique used to functionally characterize an RNA. These techniques (most commonly antisense therapy and RNA interference) can even have therapeutic benefit to treat a wide variety of genetic or infectious diseases as evidenced by the several RNA knockdown reagents currently in clinical trials. This protocol describes the use of validated gapmer antisense oligonucleotides (ASOs) to knockdown human MALAT1, a nuclear-retained lncRNA that is upregulated in multiple cancer cells. Methods used include cationic lipid transfection into HeLa cells, RNA isolation, and RT-qPCR analysis of the RNA knockdown levels.


Asunto(s)
Técnicas de Silenciamiento del Gen/métodos , Oligonucleótidos Antisentido/genética , ARN Largo no Codificante/genética , Técnicas de Silenciamiento del Gen/normas , Silenciador del Gen/fisiología , Células HeLa , Humanos , Oligonucleótidos Antisentido/farmacología , Reacción en Cadena de la Polimerasa/métodos , Interferencia de ARN/fisiología , ARN Largo no Codificante/aislamiento & purificación , Transfección/métodos
12.
Methods Mol Biol ; 2115: 23-55, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32006393

RESUMEN

Chemically modified oligonucleotides (ONs) are routinely used in the laboratory to assess gene function, and clinical advances are rapidly progressing as continual efforts are being made to optimize ON efficacy. Over the years, RNA interference (RNAi) has become one of the main tools used to inhibit RNA expression across a wide variety of species. Efforts have been made to improve the exogenous delivery of the double-stranded RNA components to the endogenous intracellular RNAi machinery to direct efficacious degradation of a user-defined RNA target. More recently, synthetic RNA ONs are being used to mimic the bacterial-derived CRISPR/Cas system to direct specific editing of the mammalian genome. Both of these techniques rely on the use of various chemical modifications to the RNA phosphate backbone or sugar in specific positions throughout the ONs to improve the desired biological outcome. Relevant chemical modifications also include conjugated targeting ligands to assist ON delivery to specific cell types. Chemical modifications are most beneficial for therapeutically relevant ONs, as they serve to enhance target binding, increase drug longevity, facilitate cell-specific targeting, improve internalization into productive intracellular compartments, and mitigate both sequence-specific as well as immune-related off-target effects (OTEs). The knowledge gained from years of optimizing RNAi reagents and characterizing the biochemical and biophysical properties of each chemical modification will hopefully accelerate the CRISPR/Cas technology into the clinic, as well as further expand the use of RNAi to treat currently undruggable diseases. This review discusses the most commonly employed chemical modifications in RNAi reagents and CRISPR/Cas guide RNAs and provides an overview of select publications that have demonstrated success in improving ON efficacy and/or mitigating undesired OTEs.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica/métodos , Interferencia de ARN , Animales , Humanos , Indicadores y Reactivos , Oligonucleótidos/química , Oligonucleótidos/genética , ARN Guía de Kinetoplastida/química , ARN Guía de Kinetoplastida/genética , ARN Interferente Pequeño/química , ARN Interferente Pequeño/genética
13.
Mol Ther ; 16(7): 1331-9, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18523447

RESUMEN

RNA interference (RNAi) is gaining acceptance as a potential therapeutic strategy against peripheral disease, and several clinical trials are already underway with 21-mer small-interfering RNA (siRNA) as the active pharmaceutical agent. However, for central affliction like pain, such innovating therapies are limited but nevertheless crucial to improve pain research and management. We demonstrate here the proof-of-concept of the use of 27-mer Dicer-substrate siRNA (DsiRNA) for silencing targets related to CNS disorders such as pain states. Indeed, low dose DsiRNA (0.005 mg/kg) was highly efficient in reducing the expression of the neurotensin receptor-2 (NTS2, a G-protein-coupled receptor (GPCR) involved in ascending nociception) in rat spinal cord through intrathecal (IT) administration formulated with the cationic lipid i-Fect. Along with specific decrease in NTS2 mRNA and protein, our results show a significant alteration in the analgesic effect of a selective-NTS2 agonist, reaching 93% inhibition up to 3-4 days after administration of DsiRNA. In order to ensure that these findings were not biased by unsuspected off-target effects (OTEs), we also demonstrated that treatment with a second NTS2-specific DsiRNA also reversed NTS2-induced antinociception, and that NTS2-specific 27-mer duplexes did not alter signaling through NTS1, a closely related receptor. Altogether, DsiRNAi represents a potent tool for dissecting nociceptive pathways and could further lead to a new class of central active drugs.


Asunto(s)
Dolor/tratamiento farmacológico , Interferencia de ARN , ARN Interferente Pequeño/uso terapéutico , Receptores de Neurotensina/antagonistas & inhibidores , Ribonucleasa III/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Ganglios Espinales/metabolismo , Masculino , Oligonucleótidos/administración & dosificación , Oligonucleótidos/uso terapéutico , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley
14.
Sci Rep ; 8(1): 15841, 2018 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-30367171

RESUMEN

Recognition of nucleic acids by endosomal Toll-like receptors (TLR) is essential to combat pathogens, but requires strict control to limit inflammatory responses. The mechanisms governing this tight regulation are unclear. We found that single-stranded oligonucleotides (ssON) inhibit endocytic pathways used by cargo destined for TLR3/4/7 signaling endosomes. Both ssDNA and ssRNA conferred the endocytic inhibition, it was concentration dependent, and required a certain ssON length. The ssON-mediated inhibition modulated signaling downstream of TLRs that localized within the affected endosomal pathway. We further show that injection of ssON dampens dsRNA-mediated inflammatory responses in the skin of non-human primates. These studies reveal a regulatory role for extracellular ssON in the endocytic uptake of TLR ligands and provide a mechanistic explanation of their immunomodulation. The identified ssON-mediated interference of endocytosis (SOMIE) is a regulatory process that temporarily dampens TLR3/4/7 signaling, thereby averting excessive immune responses.


Asunto(s)
Clatrina/metabolismo , Endocitosis/efectos de los fármacos , Oligonucleótidos/farmacología , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 7/metabolismo , Animales , Células Cultivadas , Quimiocinas/metabolismo , Citocinas/metabolismo , ADN de Cadena Simple/farmacología , Endosomas/metabolismo , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Macaca fascicularis , Poli I-C/farmacología , Transducción de Señal/efectos de los fármacos , Piel/metabolismo , Piel/patología , Receptor Toll-Like 3/antagonistas & inhibidores , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 7/antagonistas & inhibidores
15.
Methods Mol Biol ; 1517: 51-69, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27924473

RESUMEN

MicroRNAs (miRNAs) are important modulators of gene expression. Synthetic anti-microRNA oligonucleotides (AMOs, or anti-miRs) are a form of steric-blocking antisense oligonucleotides (ASOs) that inhibit miRNA function through high-affinity binding and subsequent inactivation and/or degradation of the targeted miRNA. AMOs are a primary tool used to empirically determine the biological targets of a miRNA and can also be used therapeutically when overexpression of a miRNA contributes to a disease state. Chemical modification of synthetic AMOs enhance potency by protecting the oligonucleotide from nuclease degradation and by increasing binding affinity to the target miRNA. A new steric-blocking ASO modification strategy with favorable properties for use in AMOs was recently developed that combines use of high-affinity 2'-O-methyl RNA with terminally positioned non-nucleotide "ZEN" modifiers. This protocol describes use of ZEN AMOs in a dual-luciferase reporter assay as a simplified means to validate AMO performance or to quickly test putative miRNA binding sites in target sequences. This protocol also describes a method using Western blot analysis for quantifying the level of upregulation of proteins made from an mRNA that is thought to be under miRNA regulation, following inhibition of that miRNA by ZEN AMO treatment.


Asunto(s)
MicroARNs/genética , Biología Molecular/métodos , Oligonucleótidos Antisentido/genética , Sitios de Unión , Regulación de la Expresión Génica/genética , Genes Reporteros/genética , Humanos , Luciferasas/genética , MicroARNs/antagonistas & inhibidores , MicroARNs/uso terapéutico , Oligonucleótidos Antisentido/síntesis química
16.
Oligonucleotides ; 16(1): 26-42, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16584293

RESUMEN

A wide variety of modified oligonucleotides have been tested as antisense agents. Each chemical modification produces a distinct profile of potency, toxicity, and specificity. Novel cationic phosphoramidate-modified antisense oligonucleotides have been developed recently that have unique and interesting properties. We compared the relative potency and specificity of a variety of established antisense oligonucleotides, including phosphorothioates (PS), 2'-O-methyl (2'OMe) RNAs, locked nucleic acids (LNAs), and neutral methoxyethyl (MEA) phosphoramidates with new cationic N,N-dimethylethylenediamine (DMED) phosphoramidate-modified antisense oligonucleotides. A series of oligonucleotides was synthesized that targeted two sites in the Xenopus laevis survivin gene and were introduced into Xenopus embryos by microinjection. Effects on survivin gene expression were examined using quantitative real-time PCR. Of the various modified oligonucleotide designs tested, LNA/PS chimeras (which showed the highest melting temperature) and DMED/phosphodiester chimeras (which showed protection of neighboring phosphate bonds) were potent in reducing gene expression. At 40 nM, overall specificity was superior for the LNA/PS-modified compounds compared with the DMED-modified oligonucleotides. However, at 400 nM, both of these compounds led to significant degradation of survivin mRNA, even when up to three mismatches were present in the heteroduplex.


Asunto(s)
Etilenodiaminas/química , Oligonucleótidos Antisentido/farmacología , ARN Mensajero/antagonistas & inhibidores , Proteínas de Xenopus/antagonistas & inhibidores , Amidas/química , Animales , Disparidad de Par Base , Secuencia de Bases , Desoxirribonucleasas/química , Embrión no Mamífero/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Oligonucleótidos Antisentido/química , Oligonucleótidos Antisentido/genética , Ácidos Fosfóricos/química , Survivin , Temperatura , Proteínas de Xenopus/genética , Xenopus laevis
17.
Mol Oncol ; 10(5): 693-703, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26809501

RESUMEN

Metastasis is the primary cause of death in prostate cancer (PCa) patients. Small nucleolar RNAs (snoRNAs) have long been considered "housekeeping" genes with no relevance for cancer biology. Emerging evidence has challenged this assumption, suggesting that snoRNA expression is frequently modulated during cancer progression. Despite this, no study has systematically addressed the prognostic and functional significance of snoRNAs in PCa. We performed RNA Sequencing on paired metastatic/non-metastatic PCa xenografts derived from clinical specimens. The clinical significance of differentially expressed snoRNAs was further investigated in two independent primary PCa cohorts (131 and 43 patients, respectively). The snoRNA demonstrating the strongest association with clinical outcome was quantified in PCa patient-derived serum samples and its functional relevance was investigated in PCa cells via gene expression profiling, pathway analysis and gene silencing. Our comparison revealed 21 differentially expressed snoRNAs in the metastatic vs. non-metastatic xenografts. Of those, 12 were represented in clinical databases and were further analyzed. SNORA55 emerged as a predictor of shorter relapse-free survival (results confirmed in two independent databases). SNORA55 was reproducibly detectable in serum samples from PCa patients. SNORA55 silencing in PCa cell lines significantly inhibited cell proliferation and migration. Pathway analysis revealed that SNORA55 expression is significantly associated with growth factor signaling and pro-inflammatory cytokine expression in PCa. Our results demonstrate that SNORA55 up-regulation predicts PCa progression and that silencing this non-coding gene affects PCa cell proliferation and metastatic potential, thus positioning it as both a novel biomarker and therapeutic target.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Próstata/patología , Neoplasias de la Próstata/genética , ARN Nucleolar Pequeño/genética , Transcriptoma , Anciano , Línea Celular Tumoral , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Próstata/metabolismo , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/patología , Regulación hacia Arriba
18.
PLoS One ; 10(10): e0139504, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26436892

RESUMEN

Antisense oligonucleotides (ASOs) are synthetic, single-strand RNA-DNA hybrids that induce catalytic degradation of complementary cellular RNAs via RNase H. ASOs are widely used as gene knockdown reagents in tissue culture and in Xenopus and mouse model systems. To test their effectiveness in zebrafish, we targeted 20 developmental genes and compared the morphological changes with mutant and morpholino (MO)-induced phenotypes. ASO-mediated transcript knockdown reproduced the published loss-of-function phenotypes for oep, chordin, dnd, ctnnb2, bmp7a, alk8, smad2 and smad5 in a dosage-sensitive manner. ASOs knocked down both maternal and zygotic transcripts, as well as the long noncoding RNA (lncRNA) MALAT1. ASOs were only effective within a narrow concentration range and were toxic at higher concentrations. Despite this drawback, quantitation of knockdown efficiency and the ability to degrade lncRNAs make ASOs a useful knockdown reagent in zebrafish.


Asunto(s)
Técnicas de Silenciamiento del Gen , Oligonucleótidos Antisentido/genética , ARN Mensajero/antagonistas & inhibidores , Pez Cebra/genética , Animales , Desarrollo Embrionario/genética , Estudios de Factibilidad , Femenino , Masculino , Morfolinos/genética , Morfolinos/farmacología , Oligonucleótidos Antisentido/farmacología , ARN Largo no Codificante/genética , ARN Mensajero/genética , Transcripción Genética , Pez Cebra/embriología , Proteínas de Pez Cebra/genética , Cigoto
19.
Mol Ther Nucleic Acids ; 3: e212, 2014 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-25423116

RESUMEN

Splice switching oligonucleotides (SSOs) induce alternative splicing of pre-mRNA and typically employ chemical modifications to increase nuclease resistance and binding affinity to target pre-mRNA. Here we describe a new SSO non-base modifier (a naphthyl-azo group, "ZEN™") to direct exon exclusion in mutant dystrophin pre-mRNA to generate functional dystrophin protein. The ZEN modifier is placed near the ends of a 2'-O-methyl (2'OMe) oligonucleotide, increasing melting temperature and potency over unmodified 2'OMe oligonucleotides. In cultured H2K cells, a ZEN-modified 2'OMe phosphorothioate (PS) oligonucleotide delivered by lipid transfection greatly enhanced dystrophin exon skipping over the same 2'OMePS SSO lacking ZEN. However, when tested using free gymnotic uptake in vitro and following systemic delivery in vivo in dystrophin deficient mdx mice, the same ZEN-modified SSO failed to enhance potency. Importantly, we show for the first time that in vivo activity of anionic SSOs is modelled in vitro only when using gymnotic delivery. ZEN is thus a novel modifier that enhances activity of SSOs in vitro but will require improved delivery methods before its in vivo clinical potential can be realized.

20.
Mol Ther Nucleic Acids ; 2: e67, 2013 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-23299833

RESUMEN

The physiological barriers of the brain impair drug delivery for treatment of many neurological disorders. One delivery approach that has not been investigated for their ability to penetrate the brain is RNA-based aptamers. These molecules can impart delivery to peripheral tissues and circulating immune cells, where they act as ligand mimics or can be modified to carry payloads. We developed a library of aptamers and an in vivo evolution protocol to determine whether specific aptamers could be identified that would home to the brain after injection into the peripheral vasculature. Unlike biopanning with recombinant bacteriophage libraries, we found that the aptamer library employed here required more than 15 rounds of in vivo selection for convergence to specific sequences. The aptamer species identified through this approach bound to brain capillary endothelia and penetrated into the parenchyma. The methods described may find general utility for targeting various payloads to the brain.Molecular Therapy - Nucleic Acids (2013) 2, e67; doi:10.1038/mtna.2012.59; published online 8 January 2013.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA