Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Type of study
Publication year range
1.
Nucleic Acid Ther ; 30(1): 4-13, 2020 02.
Article in English | MEDLINE | ID: mdl-31618108

ABSTRACT

Methods for the quantification of antisense oligonucleotides (AONs) provide insightful information on biodistribution and intracellular trafficking. However, the established methods have not provided information on the absolute number of molecules in subcellular compartments or about how many AONs are needed for target gene reduction for unconjugated AONs. We have developed a new method for nuclear AON quantification that enables us to determine the absolute number of AONs per nucleus without relying on AON conjugates such as fluorophores that may alter AON distribution. This study describes an alternative and label-free method using subcellular fractionation, nucleus counting, and locked nucleic acid (LNA) sandwich enzyme-linked immunosorbent assay to quantify absolute numbers of oligonucleotides in nuclei. Our findings show compound variability (diversity) by which 247,000-693,000 LNAs/nuclei results in similar target reduction for different compounds. This method can be applied to any antisense drug discovery platform providing information on specific and clinically relevant AONs. Finally, this method can directly compare nuclear entry of AON with target gene knockdown for any compound design and nucleobase sequence, gene target, and phosphorothioate stereochemistry.


Subject(s)
Molecular Targeted Therapy , Oligonucleotides, Antisense/isolation & purification , Oligonucleotides/isolation & purification , Tissue Distribution/genetics , Cell Nucleus/genetics , Enzyme-Linked Immunosorbent Assay/methods , Humans , Oligonucleotides/therapeutic use , Oligonucleotides, Antisense/therapeutic use , Tissue Distribution/drug effects
2.
Mol Ther Nucleic Acids ; 8: 158-168, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28918018

ABSTRACT

Two mechanisms dominate the clinical pipeline for oligonucleotide-based gene silencing, namely, the antisense approach that recruits RNase H to cleave target RNA and the RNAi approach that recruits the RISC complex to cleave target RNA. Multiple chemical designs can be used to elicit each pathway. We compare the silencing of the asthma susceptibility gene ADAM33 in MRC-5 lung fibroblasts using four classes of gene silencing agents, two that use each mechanism: traditional duplex small interfering RNAs (siRNAs), single-stranded small interfering RNAs (ss-siRNAs), locked nucleic acid (LNA) gapmer antisense oligonucleotides (ASOs), and novel hexadecyloxypropyl conjugates of the ASOs. Of these designs, the gapmer ASOs emerged as lead compounds for silencing ADAM33 expression: several gapmer ASOs showed subnanomolar potency when transfected with cationic lipid and low micromolar potency with no toxicity when delivered gymnotically. The preferential susceptibility of ADAM33 mRNA to silencing by RNase H may be related to the high degree of nuclear retention observed for this mRNA. Dynamic light scattering data showed that the hexadecyloxypropyl ASO conjugates self-assemble into clusters. These conjugates showed reduced potency relative to unconjugated ASOs unless the lipophilic tail was conjugated to the ASO using a biocleavable linkage. Finally, based on the lead ASOs from (human) MRC-5 cells, we developed a series of homologous ASOs targeting mouse Adam33 with excellent activity. Our work confirms that ASO-based gene silencing of ADAM33 is a useful tool for asthma research and therapy.

3.
Nucleic Acid Ther ; 26(4): 216-22, 2016 08.
Article in English | MEDLINE | ID: mdl-27123752

ABSTRACT

Single-stranded silencing RNAs (ss-siRNAs) are chemically modified single-stranded oligomers that engage the RNA interference machinery normally used by duplex RNAs to silence gene expression. ss-siRNAs have the potential to combine advantages of antisense oligonucleotides and siRNAs. Previous work has explored the chemistry of the phosphate and the oligonucleotide body. We now describe the process of attempting to develop and optimize ss-siRNAs based on five active siRNA duplexes. Three of the sequences failed to show any activity as ss-siRNAs, and in two of those cases the ss-siRNAs showed significantly increased toxicity relative to the parent duplexes. For the two sequences that did work well as ss-siRNAs, we show that the chemistry of the 3'-terminal dinucleotide also has a significant effect on the potency of ss-siRNAs. Previously published work on ss-siRNAs has been based on a 2'-O-methoxyethyl-RNA (MOE) dinucleotide at the 3'-terminus. To our surprise, oligomers containing 2'-O-Me-RNA modifications at the 3'-terminus showed significantly improved potency and activity relative to those modified with MOE at the same sites. Oligonucleotides with two locked nucleic acid units at the 3'-terminus showed improved activity over the MOE-modified analog for one sequence. Importantly, the fact that 2'-O-Me-RNA works so well makes the ss-siRNA approach accessible to a wider range of researchers since it can be achieved with inexpensive commercially available modifications.


Subject(s)
Gene Silencing , Oligonucleotides, Antisense/genetics , RNA Interference , RNA, Small Interfering/genetics , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/genetics , Humans , MCF-7 Cells , Oligonucleotides, Antisense/therapeutic use , RNA, Small Interfering/therapeutic use
4.
Adv Mater ; 27(4): 750-8, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25413444

ABSTRACT

A strategy to exploit aptamers as recognition elements of molecularly imprinted polymeric nanoparticles (AptaMIP NPs) is presented, via modification of the chemical structure of the DNA. It is demonstrated that the introduction of this modified "aptamer monomer" results in an increase of the affinity of the produced MIP NPs, without altering their physical properties such as size, shape, or dispersibility.


Subject(s)
Aptamers, Nucleotide/chemistry , Molecular Imprinting , Nanoparticles/chemistry , Polymers/chemistry , Polymers/chemical synthesis , Aptamers, Nucleotide/genetics , Base Sequence , Models, Molecular , Molecular Conformation
5.
Nucleic Acid Ther ; 24(3): 199-209, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24694346

ABSTRACT

Mutant huntingtin (HTT) protein is the cause of Huntington's disease (HD), an incurable neurological disorder. Almost all patients are heterozygous for mutant HTT and approaches that reduce levels of mutant HTT while leaving expression of wild-type HTT intact might be ideal options for therapeutic development. We have developed several allele-selective strategies for silencing HTT, including single-stranded silencing RNAs (ss-siRNAs). ss-siRNAs are oligonucleotides containing chemical modifications that permit action through the RNA interference (RNAi) pathway. Modified ss-siRNAs chosen to test the effects of varying oligomer length, lipid modification, the introduction of mismatched bases, and variation of chemical modification. We find that several modified ss-siRNA are potent and allele-selective inhibitors of HTT expression. An ss-siRNA with three mismatched bases relative to the CAG repeat was an allele-selective inhibitor of HTT expression in the HdhQ175 mouse model. Multiple allele-selective ss-siRNAs provide a wide platform of modifications to draw on for further optimization and therapeutic development. Our data provide insights into how ss-siRNAs can be modified to improve their properties and facilitate the discovery of the lead compounds necessary for further development.


Subject(s)
Alleles , Brain/metabolism , Huntington Disease/genetics , Nerve Tissue Proteins/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Animals , Base Sequence , Brain/pathology , Cell Line , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Humans , Huntingtin Protein , Huntington Disease/metabolism , Huntington Disease/pathology , Injections, Intraventricular , Lipids/chemistry , Mice , Molecular Sequence Data , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , RNA Interference , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , RNA, Small Interfering/chemical synthesis , RNA, Small Interfering/metabolism , Structure-Activity Relationship
6.
Nucleic Acids Res ; 41(20): 9570-83, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23935115

ABSTRACT

Single-stranded silencing RNAs (ss-siRNAs) provide an alternative approach to gene silencing. ss-siRNAs combine the simplicity and favorable biodistribution of antisense oligonucleotides with robust silencing through RNA interference (RNAi). Previous studies reported potent and allele-selective inhibition of human huntingtin expression by ss-siRNAs that target the expanded CAG repeats within the mutant allele. Mutant ataxin-3, the genetic cause of Machado-Joseph Disease, also contains an expanded CAG repeat. We demonstrate here that ss-siRNAs are allele-selective inhibitors of ataxin-3 expression and then redesign ss-siRNAs to optimize their selectivity. We find that both RNAi-related and non-RNAi-related mechanisms affect gene expression by either blocking translation or affecting alternative splicing. These results have four broad implications: (i) ss-siRNAs will not always behave similarly to analogous RNA duplexes; (ii) the sequences surrounding CAG repeats affect allele-selectivity of anti-CAG oligonucleotides; (iii) ss-siRNAs can function through multiple mechanisms and; and (iv) it is possible to use chemical modification to optimize ss-siRNA properties and improve their potential for drug discovery.


Subject(s)
Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , RNA Interference , RNA, Small Interfering/chemistry , Repressor Proteins/genetics , Alleles , Alternative Splicing , Amino Acid Sequence , Ataxin-3 , Cell Line , Humans , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nuclear Proteins/chemistry , Repressor Proteins/chemistry , Trinucleotide Repeats
7.
Nucleic Acids Res ; 41(18): 8788-801, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23887934

ABSTRACT

Abasic substitutions within DNA or RNA are tools for evaluating the impact of absent nucleobases. Because of the importance of abasic sites in genetic damage, most research has involved DNA. Little information is available on the impact of abasic substitutions within RNA or on RNA interference (RNAi). Here, we examine the effect of abasic substitutions on RNAi and allele-selective gene silencing. Huntington's disease (HD) and Machado Joseph Disease (MJD) are severe neurological disorders that currently have no cure. HD and MJD are caused by an expansion of CAG repeats within one mRNA allele encoding huntingtin (HTT) and ataxin-3 (ATX-3) proteins. Agents that silence mutant HTT or ATX-3 expression would remove the cause of HD or MJD and provide an option for therapeutic development. We describe flexible syntheses for abasic substitutions and show that abasic RNA duplexes allele-selectively inhibit both mutant HTT and mutant ATX-3. Inhibition involves the RNAi protein argonaute 2, even though the abasic substitution disrupts the catalytic cleavage of RNA target by argonaute 2. Several different abasic duplexes achieve potent and selective inhibition, providing a broad platform for subsequent development. These findings introduce abasic substitutions as a tool for tailoring RNA duplexes for gene silencing.


Subject(s)
Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , RNA Interference , RNA, Small Interfering/chemistry , Repressor Proteins/genetics , Alleles , Ataxin-3 , Base Pair Mismatch , Cell Line , Humans , Huntingtin Protein , Huntington Disease/genetics , Machado-Joseph Disease/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , RNA, Small Interfering/chemical synthesis , Repressor Proteins/metabolism
8.
Cell ; 150(5): 895-908, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22939619

ABSTRACT

Mutant huntingtin (HTT) protein causes Huntington disease (HD), an incurable neurological disorder. Silencing mutant HTT using nucleic acids would eliminate the root cause of HD. Developing nucleic acid drugs is challenging, and an ideal clinical approach to gene silencing would combine the simplicity of single-stranded antisense oligonucleotides with the efficiency of RNAi. Here, we describe RNAi by single-stranded siRNAs (ss-siRNAs). ss-siRNAs are potent (>100-fold more than unmodified RNA) and allele-selective (>30-fold) inhibitors of mutant HTT expression in cells derived from HD patients. Strategic placement of mismatched bases mimics micro-RNA recognition and optimizes discrimination between mutant and wild-type alleles. ss-siRNAs require Argonaute protein and function through the RNAi pathway. Intraventricular infusion of ss-siRNA produced selective silencing of the mutant HTT allele throughout the brain in a mouse HD model. These data demonstrate that chemically modified ss-siRNAs function through the RNAi pathway and provide allele-selective compounds for clinical development.


Subject(s)
Disease Models, Animal , Huntington Disease/genetics , Huntington Disease/therapy , Nerve Tissue Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Animals , Brain/metabolism , Cell Line , Humans , Huntingtin Protein , Mice , Oligodeoxyribonucleotides, Antisense/genetics , RNA, Small Interfering/genetics
9.
J Am Chem Soc ; 133(22): 8404-7, 2011 Jun 08.
Article in English | MEDLINE | ID: mdl-21539318

ABSTRACT

Oligonucleotides and their derivatives are a proven chemical strategy for modulating gene expression. However, their negative charge remains a challenge for delivery and target recognition inside cells. Here we show that oligonucleotide-oligospermine conjugates (Zip nucleic acids or ZNAs) can help overcome these shortcomings by serving as effective antisense and antigene agents. Conjugates containing DNA and locked nucleic acid (LNA) oligonucleotides are active, and oligospermine conjugation facilitates carrier-free cell uptake at nanomolar concentrations. Conjugates targeting the CAG triplet repeat within huntingtin (HTT) mRNA selectively inhibit expression of the mutant huntingtin protein. Conjugates targeting the promoter of the progesterone receptor (PR) function as antigene agents to block PR expression. These observations support further investigation of ZNA conjugates as gene silencing agents.


Subject(s)
Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Oligonucleotides, Antisense/antagonists & inhibitors , Oligonucleotides/chemistry , Oligonucleotides/pharmacology , Spermine/chemistry , Base Sequence , Cations , Cell Membrane Permeability/drug effects , Drug Delivery Systems , Gene Transfer Techniques , Inhibitory Concentration 50 , Models, Biological , Models, Molecular , Molecular Sequence Data , Molecular Structure , Oligonucleotides/metabolism , Spermine/metabolism , Spermine/pharmacology
10.
Biochemistry ; 49(47): 10166-78, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-21028906

ABSTRACT

Huntington's disease (HD) is a currently incurable neurodegenerative disease caused by the expansion of a CAG trinucleotide repeat within the huntingtin (HTT) gene. Therapeutic approaches include selectively inhibiting the expression of the mutated HTT allele while conserving function of the normal allele. We have evaluated a series of antisense oligonucleotides (ASOs) targeted to the expanded CAG repeat within HTT mRNA for their ability to selectively inhibit expression of mutant HTT protein. Several ASOs incorporating a variety of modifications, including bridged nucleic acids and phosphorothioate internucleotide linkages, exhibited allele-selective silencing in patient-derived fibroblasts. Allele-selective ASOs did not affect the expression of other CAG repeat-containing genes and selectivity was observed in cell lines containing minimal CAG repeat lengths representative of most HD patients. Allele-selective ASOs left HTT mRNA intact and did not support ribonuclease H activity in vitro. We observed cooperative binding of multiple ASO molecules to CAG repeat-containing HTT mRNA transcripts in vitro. These results are consistent with a mechanism involving inhibition at the level of translation. ASOs targeted to the CAG repeat of HTT provide a starting point for the development of oligonucleotide-based therapeutics that can inhibit gene expression with allelic discrimination in patients with HD.


Subject(s)
Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Oligonucleotides, Antisense/pharmacology , Alleles , Cell Line , Gene Expression/drug effects , Humans , Huntingtin Protein , Huntington Disease/drug therapy , Huntington Disease/genetics , Oligonucleotides/genetics , Oligonucleotides/pharmacology , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/therapeutic use , Phosphorothioate Oligonucleotides/pharmacology , Ribonuclease H/metabolism , Structure-Activity Relationship , Trinucleotide Repeat Expansion/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...