Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nucleic Acids Res ; 49(16): 9026-9041, 2021 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-34417625

RESUMEN

The PS modification enhances the nuclease stability and protein binding properties of gapmer antisense oligonucleotides (ASOs) and is one of very few modifications that support RNaseH1 activity. We evaluated the effect of introducing stereorandom and chiral mesyl-phosphoramidate (MsPA) linkages in the DNA gap and flanks of gapmer PS ASOs and characterized the effect of these linkages on RNA-binding, nuclease stability, protein binding, pro-inflammatory profile, antisense activity and toxicity in cells and in mice. We show that all PS linkages in a gapmer ASO can be replaced with MsPA without compromising chemical stability and RNA binding affinity but these designs reduced activity. However, replacing up to 5 PS in the gap with MsPA was well tolerated and replacing specific PS linkages at appropriate locations was able to greatly reduce both immune stimulation and cytotoxicity. The improved nuclease stability of MsPA over PS translated to significant improvement in the duration of ASO action in mice which was comparable to that of enhanced stabilized siRNA designs. Our work highlights the combination of PS and MsPA linkages as a next generation chemical platform for identifying ASO drugs with improved potency and therapeutic index, reduced pro-inflammatory effects and extended duration of effect.


Asunto(s)
Oligonucleótidos Antisentido/síntesis química , Índice Terapéutico de los Medicamentos , Animales , Células HEK293 , Células HeLa , Humanos , Hígado/metabolismo , Masculino , Mesilatos/química , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Oligonucleótidos Antisentido/farmacocinética , Oligonucleótidos Antisentido/toxicidad , Fosforamidas/química , Unión Proteica , Distribución Tisular
2.
Nucleic Acids Res ; 48(17): 9840-9858, 2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32870273

RESUMEN

Antisense oligonucleotide (ASO) drugs that trigger RNase H1 cleavage of target RNAs have been developed to treat various diseases. Basic pharmacological principles suggest that the development of tolerance is a common response to pharmacological interventions. In this manuscript, for the first time we report a molecular mechanism of tolerance that occurs with some ASOs. Two observations stimulated our interest: some RNA targets are difficult to reduce with RNase H1 activating ASOs and some ASOs display a shorter duration of activity than the prolonged target reduction typically observed. We found that certain ASOs targeting the coding region of some mRNAs that initially reduce target mRNAs can surprisingly increase the levels of the corresponding pre-mRNAs. The increase in pre-mRNA is delayed and due to enhanced transcription and likely also slower processing. This process requires that the ASOs bind in the coding region and reduce the target mRNA by RNase H1 while the mRNA resides in the ribosomes. The pre-mRNA increase is dependent on UPF3A and independent of the NMD pathway or the XRN1-CNOT pathway. The response is consistent in multiple cell lines and independent of the methods used to introduce ASOs into cells.


Asunto(s)
Oligonucleótidos Antisentido/genética , ARN Mensajero/genética , Ribonucleasa H/metabolismo , Animales , Emparejamiento Base , Células HEK293 , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Oligonucleótidos Antisentido/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo
3.
Nucleic Acids Res ; 48(3): 1372-1391, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-31840180

RESUMEN

Release of phosphorothioate antisense oligonucleotides (PS-ASOs) from late endosomes (LEs) is a rate-limiting step and a poorly defined process for productive intracellular ASO drug delivery. Here, we examined the role of Golgi-endosome transport, specifically M6PR shuttling mediated by GCC2, in PS-ASO trafficking and activity. We found that reduction in cellular levels of GCC2 or M6PR impaired PS-ASO release from endosomes and decreased PS-ASO activity in human cells. GCC2 relocated to LEs upon PS-ASO treatment, and M6PR also co-localized with PS-ASOs in LEs or on LE membranes. These proteins act through the same pathway to influence PS-ASO activity, with GCC2 action preceding that of M6PR. Our data indicate that M6PR binds PS-ASOs and facilitates their vesicular escape. The co-localization of M6PR and of GCC2 with ASOs is influenced by the PS modifications, which have been shown to enhance the affinity of ASOs for proteins, suggesting that localization of these proteins to LEs is mediated by ASO-protein interactions. Reduction of M6PR levels also decreased PS-ASO activity in mouse cells and in livers of mice treated subcutaneously with PS-ASO, indicating a conserved mechanism. Together, these results demonstrate that the transport machinery between LE and Golgi facilitates PS-ASO release.


Asunto(s)
Endosomas/genética , Proteínas de la Matriz de Golgi/genética , Oligonucleótidos Antisentido/genética , Receptor IGF Tipo 2/genética , Animales , Endocitosis/genética , Endosomas/metabolismo , Aparato de Golgi/genética , Aparato de Golgi/metabolismo , Proteínas de la Matriz de Golgi/metabolismo , Células HeLa , Humanos , Ratones , Oligonucleótidos Fosforotioatos/genética , Transporte de Proteínas/genética , Receptor IGF Tipo 2/metabolismo
4.
Nucleic Acids Res ; 48(4): 1691-1700, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-31980820

RESUMEN

Therapeutic oligonucleotides are often modified using the phosphorothioate (PS) backbone modification which enhances stability from nuclease mediated degradation. However, substituting oxygen in the phosphodiester backbone with sulfur introduce chirality into the backbone such that a full PS 16-mer oligonucleotide is comprised of 215 distinct stereoisomers. As a result, the role of PS chirality on the performance of antisense oligonucleotides (ASOs) has been a subject of debate for over two decades. We carried out a systematic analysis to determine if controlling PS chirality in the DNA gap region can enhance the potency and safety of gapmer ASOs modified with high-affinity constrained Ethyl (cEt) nucleotides in the flanks. As part of this effort, we examined the effect of systematically controlling PS chirality on RNase H1 cleavage patterns, protein mislocalization phenotypes, activity and toxicity in cells and in mice. We found that while controlling PS chirality can dramatically modulate interactions with RNase H1 as evidenced by changes in RNA cleavage patterns, these were insufficient to improve the overall therapeutic profile. We also found that controlling PS chirality of only two PS linkages in the DNA gap was sufficient to modulate RNase H1 cleavage patterns and combining these designs with simple modifications such as 2'-OMe to the DNA gap resulted in dramatic improvements in therapeutic index. However, we were unable to demonstrate improved potency relative to the stereorandom parent ASO or improved safety over the 2'-OMe gap-modified stereorandom parent ASO. Overall, our work shows that while controlling PS chirality can modulate RNase H1 cleavage patterns, ASO sequence and design are the primary drivers which determine the pharmacological and toxicological properties of gapmer ASOs.


Asunto(s)
ADN/genética , Oligonucleótidos Antisentido/genética , Oligonucleótidos Fosforotioatos/genética , Ribonucleasa H/genética , Animales , ADN/química , Ratones , Oligonucleótidos Antisentido/química , Oligonucleótidos Fosforotioatos/química , Unión Proteica/genética , Ribonucleasa H/química
5.
Nucleic Acids Res ; 47(11): 5465-5479, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31034558

RESUMEN

Phosphorothioate-modified antisense oligonucleotides (PS-ASOs) interact with a host of plasma, cell-surface and intracellular proteins which govern their therapeutic properties. Given the importance of PS backbone for interaction with proteins, we systematically replaced anionic PS-linkages in toxic ASOs with charge-neutral alkylphosphonate linkages. Site-specific incorporation of alkyl phosphonates altered the RNaseH1 cleavage patterns but overall rates of cleavage and activity versus the on-target gene in cells and in mice were only minimally affected. However, replacing even one PS-linkage at position 2 or 3 from the 5'-side of the DNA-gap with alkylphosphonates reduced or eliminated toxicity of several hepatotoxic gapmer ASOs. The reduction in toxicity was accompanied by the absence of nucleolar mislocalization of paraspeckle protein P54nrb, ablation of P21 mRNA elevation and caspase activation in cells, and hepatotoxicity in mice. The generality of these observations was further demonstrated for several ASOs versus multiple gene targets. Our results add to the types of structural modifications that can be used in the gap-region to enhance ASO safety and provide insights into understanding the biochemistry of PS ASO protein interactions.


Asunto(s)
Membrana Celular/metabolismo , Citoplasma/metabolismo , Oligonucleótidos Antisentido/química , Organofosfonatos/química , Oligonucleótidos Fosforotioatos/química , Células 3T3-L1 , Animales , Caspasas/metabolismo , Línea Celular , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Proteínas de Unión al ADN , Células HeLa , Hepatocitos/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas Asociadas a Matriz Nuclear/metabolismo , Factores de Transcripción de Octámeros/genética , Factores de Transcripción de Octámeros/metabolismo , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos Fosforotioatos/administración & dosificación , Unión Proteica , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ribonucleasa H/genética , Ribonucleasa H/metabolismo , Receptores Depuradores de Clase B/genética , Receptores Depuradores de Clase B/metabolismo
6.
Nucleic Acids Res ; 46(5): 2204-2217, 2018 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-29390093

RESUMEN

We reported previously that a 2' fluoro-modified (2' F) phosphorothioate (PS) antisense oligonucleotides (ASOs) with 5-10-5 gapmer configuration interacted with proteins from Drosophila behavior/human splicing (DBHS) family with higher affinity than PS-ASOs modified with 2'-O-(2-methoxyethyl) (2' MOE) or 2',4'-constrained 2'-O-ethyl (cEt) did. Rapid degradation of these proteins and cytotoxicity were observed in cells treated with 2' F PS-ASO. Here, we report that 2' F gapmer PS-ASOs of different sequences caused reduction in levels of DBHS proteins and hepatotoxicity in mice. 2' F PS-ASOs induced activation of the P53 pathway and downregulation of metabolic pathways. Altered levels of RNA and protein markers for hepatotoxicity, liver necrosis, and apoptosis were observed as early as 24 to 48 hours after a single administration of the 2' F PS-ASO. The observed effects were not likely due to the hybridization-dependent RNase H1 cleavage of on- or potential off-target RNAs, or due to potential toxicity of 2' F nucleoside metabolites. Instead, we found that 2' F PS-ASO associated with more intra-cellular proteins including proteins from DBHS family. Our results suggest that protein-binding correlates positively with the 2' F modification-dependent loss of DBHS proteins and the toxicity of gapmer 2' F PS-ASO in vivo.


Asunto(s)
Hepatocitos/efectos de los fármacos , Hígado/efectos de los fármacos , Oligonucleótidos Antisentido/toxicidad , Oligonucleótidos Fosforotioatos/metabolismo , Transcriptoma/efectos de los fármacos , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Hígado/metabolismo , Hígado/patología , Masculino , Ratones Endogámicos BALB C , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Factor de Empalme Asociado a PTB/genética , Factor de Empalme Asociado a PTB/metabolismo , Oligonucleótidos Fosforotioatos/genética , Unión Proteica/efectos de los fármacos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transcriptoma/genética
7.
Nucleic Acids Res ; 45(18): 10672-10692, 2017 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-28977560

RESUMEN

An R-loop is a DNA:RNA hybrid formed during transcription when a DNA duplex is invaded by a nascent RNA transcript. R-loops accumulate in nucleoli during RNA polymerase I (RNAP I) transcription. Here, we report that mammalian RNase H1 enriches in nucleoli and co-localizes with R-loops in cultured human cells. Co-migration of RNase H1 and R-loops from nucleoli to perinucleolar ring structures was observed upon inhibition of RNAP I transcription. Treatment with camptothecin which transiently stabilized nucleolar R-loops recruited RNase H1 to the nucleoli. It has been reported that the absence of Topoisomerase and RNase H activity in Escherichia coli or Saccharomyces cerevisiae caused R-loop accumulation along rDNA. We found that the distribution of RNase H1 and Top1 along rDNA coincided at sites where R-loops accumulated in mammalian cells. Loss of either RNase H1 or Top1 caused R-loop accumulation, and the accumulation of R-loops was exacerbated when both proteins were depleted. Importantly, we observed that protein levels of Top1 were negatively correlated with the abundance of RNase H1. We conclude that Top1 and RNase H1 are partially functionally redundant in mammalian cells to suppress RNAP I transcription-associate R-loops.


Asunto(s)
Nucléolo Celular/genética , ADN Ribosómico/química , ARN Polimerasa I/metabolismo , Ribonucleasa H/análisis , Transcripción Genética , Animales , Camptotecina/farmacología , Nucléolo Celular/efectos de los fármacos , Nucléolo Celular/enzimología , Daño del ADN , ADN-Topoisomerasas de Tipo I/análisis , ADN Ribosómico/metabolismo , Células HEK293 , Células HeLa , Humanos , Ratones Noqueados , Dominios Proteicos , ARN/química , ARN Polimerasa I/análisis , Ribonucleasa H/química , Ribonucleasa H/metabolismo , Transcripción Genética/efectos de los fármacos
8.
Nucleic Acids Res ; 44(7): 3351-63, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-26843429

RESUMEN

DNA-based antisense oligonucleotides (ASOs) elicit cleavage of the targeted RNA by the endoribonuclease RNase H1, whereas siRNAs mediate cleavage through the RNAi pathway. To determine the fates of the cleaved RNA in cells, we lowered the levels of the factors involved in RNA surveillance prior to treating cells with ASOs or siRNA and analyzed cleavage products by RACE. The cytoplasmic 5' to 3' exoribonuclease XRN1 was responsible for the degradation of the downstream cleavage products generated by ASOs or siRNA targeting mRNAs. In contrast, downstream cleavage products generated by ASOs targeting nuclear long non-coding RNA Malat 1 and pre-mRNA were degraded by nuclear XRN2. The downstream cleavage products did not appear to be degraded in the 3' to 5' direction as the majority of these products contained intact poly(A) tails and were bound by the poly(A) binding protein. The upstream cleavage products of Malat1 were degraded in the 3' to 5' direction by the exosome complex containing the nuclear exoribonuclease Dis3. The exosome complex containing Dis3 or cytoplasmic Dis3L1 degraded mRNA upstream cleavage products, which were not bound by the 5'-cap binding complex and, consequently, were susceptible to degradation in the 5' to 3' direction by the XRN exoribonucleases.


Asunto(s)
Exorribonucleasas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Oligonucleótidos Antisentido , División del ARN , ARN Interferente Pequeño/metabolismo , Complejo Multienzimático de Ribonucleasas del Exosoma/metabolismo , Células HeLa , Humanos , Interferencia de ARN , Procesamiento Postranscripcional del ARN , ARN Largo no Codificante/metabolismo
9.
Nat Biotechnol ; 37(6): 640-650, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31036929

RESUMEN

The molecular mechanisms of toxicity of chemically modified phosphorothioate antisense oligonucleotides (PS-ASOs) are not fully understood. Here, we report that toxic gapmer PS-ASOs containing modifications such as constrained ethyl (cEt), locked nucleic acid (LNA) and 2'-O-methoxyethyl (2'-MOE) bind many cellular proteins with high avidity, altering their function, localization and stability. We show that RNase H1-dependent delocalization of paraspeckle proteins to nucleoli is an early event in PS-ASO toxicity, followed by nucleolar stress, p53 activation and apoptotic cell death. Introduction of a single 2'-O-methyl (2'-OMe) modification at gap position 2 reduced protein-binding, substantially decreasing hepatotoxicity and improving the therapeutic index with minimal impairment of antisense activity. We validated the ability of this modification to generally mitigate PS-ASO toxicity with more than 300 sequences. Our findings will guide the design of PS-ASOs with optimal therapeutic profiles.


Asunto(s)
Oligonucleótidos Antisentido/química , Oligonucleótidos/química , Oligonucleótidos Fosforotioatos/química , Humanos , Hígado/efectos de los fármacos , Oligonucleótidos/uso terapéutico , Oligonucleótidos Antisentido/uso terapéutico , Oligonucleótidos Fosforotioatos/uso terapéutico , Unión Proteica/efectos de los fármacos , Ribonucleasa H/química , Ribonucleasa H/genética , Índice Terapéutico
10.
PLoS One ; 12(3): e0173494, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28288210

RESUMEN

Altered expression of NEAT1, the architectural long non-coding RNA (lncRNA) of nuclear paraspeckles, has been reported during tumorigenesis, as well as under various cellular stress conditions. Here we report that the depletion of NEAT1 lncRNA alleviates nucleolar stress during RNAP I inhibition through releasing sequestered P54nrb and PSF to facilitate the IRES-dependent translation of c-Myc. RNAP I inhibitor CX5461 disrupts the SL1-rDNA interaction and induces nucleolar disruption, demonstrated by the accumulation of fibrillarin-containing nucleoplasmic foci and nucleolar clearance of ribosomal proteins in HeLa cells. Antisense oligonucleotide-mediated depletion of NEAT1 lncRNA significantly attenuated the RNAP I inhibition and its related nucleolar disruption. Interestingly, induction in the levels of c-Myc protein was observed in NEAT1-depeleted cells under RNAP I inhibition. NEAT1-associated paraspeckle proteins P54nrb and PSF have been reported as positive regulators of c-Myc translation through interaction with c-Myc IRES. Indeed, an increased association of P54nrb and PSF with c-Myc mRNA was observed in NEAT1-depleted cells. Moreover, apoptosis was observed in HeLa cells depleted of P54nrb and PSF, further confirming the positive involvement of P54nrb and PSF in cell proliferation. Together, our results suggest that NEAT1 depletion rescues CX5461-induced nucleolar stress through facilitating c-Myc translation by relocating P54nrb/PSF from nuclear paraspeckles to c-Myc mRNAs.


Asunto(s)
Genes myc , Proteínas Asociadas a Matriz Nuclear/metabolismo , Factores de Transcripción de Octámeros/metabolismo , Factor de Empalme Asociado a PTB/metabolismo , Biosíntesis de Proteínas , ARN Largo no Codificante/fisiología , Proteínas de Unión al ARN/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN , Humanos , ARN Mensajero/genética , ARN Ribosómico/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA