Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 4 de 4
1.
J Biol Chem ; 298(10): 102398, 2022 10.
Article En | MEDLINE | ID: mdl-35988651

Unusual nucleic acid structures play vital roles as intermediates in many cellular processes and, in the case of peptide nucleic acid (PNA)-mediated triplexes, are leveraged as tools for therapeutic gene editing. However, due to their transient nature, an understanding of the factors that interact with and process dynamic nucleic acid structures remains limited. Here, we developed snapELISA (structure-specific nucleic acid-binding protein ELISA), a rapid high-throughput platform to interrogate and compare up to 2688 parallel nucleic acid structure-protein interactions in vitro. We applied this system to both triplex-forming oligonucleotide-induced DNA triplexes and DNA-bound PNA heterotriplexes to describe the identification of previously known and novel interactors for both structures. For PNA heterotriplex recognition analyses, snapELISA identified factors implicated in nucleotide excision repair (XPA, XPC), single-strand annealing repair (RAD52), and recombination intermediate structure binding (TOP3A, BLM, MUS81). We went on to validate selected factor localization to genome-targeted PNA structures within clinically relevant loci in human cells. Surprisingly, these results demonstrated XRCC5 localization to PNA triplex-forming sites in the genome, suggesting the presence of a double-strand break intermediate. These results describe a powerful comparative approach for identifying structure-specific nucleic acid interactions and expand our understanding of the mechanisms of triplex structure recognition and repair.


DNA , Peptide Nucleic Acids , Humans , DNA/chemistry , DNA/metabolism , Enzyme-Linked Immunosorbent Assay , Nucleic Acid Conformation , Peptide Nucleic Acids/chemistry , Peptide Nucleic Acids/metabolism
2.
Nucleic Acids Res ; 50(10): e59, 2022 06 10.
Article En | MEDLINE | ID: mdl-35235944

Despite the rapid and broad implementation of CRISPR-Cas9-based technologies, convenient tools to modulate dose, timing, and precision remain limited. Building on methods using synthetic peptide nucleic acids (PNAs) to bind RNA with unusually high affinity, we describe guide RNA (gRNA) spacer-targeted, or 'antispacer', PNAs as a tool to modulate Cas9 binding and activity in cells in a sequence-specific manner. We demonstrate that PNAs rapidly and efficiently target complexed gRNA spacer sequences at low doses and without design restriction for sequence-selective Cas9 inhibition. We further show that short PAM-proximal antispacer PNAs achieve potent cleavage inhibition (over 2000-fold reduction) and that PAM-distal PNAs modify gRNA affinity to promote on-target specificity. Finally, we apply antispacer PNAs for temporal regulation of two dCas9-fusion systems. These results present a novel rational approach to nucleoprotein engineering and describe a rapidly implementable antisense platform for CRISPR-Cas9 modulation to improve spatiotemporal versatility and safety across applications.


Peptide Nucleic Acids , RNA, Guide, Kinetoplastida , CRISPR-Cas Systems , Gene Editing/methods , Peptide Nucleic Acids/pharmacology , RNA, Guide, Kinetoplastida/genetics
3.
Nature ; 582(7813): 586-591, 2020 06.
Article En | MEDLINE | ID: mdl-32494005

Deregulation of metabolism and disruption of genome integrity are hallmarks of cancer1. Increased levels of the metabolites 2-hydroxyglutarate, succinate and fumarate occur in human malignancies owing to somatic mutations in the isocitrate dehydrogenase-1 or -2 (IDH1 or IDH2) genes, or germline mutations in the fumarate hydratase (FH) and succinate dehydrogenase genes (SDHA, SDHB, SDHC and SDHD), respectively2-4. Recent work has made an unexpected connection between these metabolites and DNA repair by showing that they suppress the pathway of homology-dependent repair (HDR)5,6 and confer an exquisite sensitivity to inhibitors of poly (ADP-ribose) polymerase (PARP) that are being tested in clinical trials. However, the mechanism by which these oncometabolites inhibit HDR remains poorly understood. Here we determine the pathway by which these metabolites disrupt DNA repair. We show that oncometabolite-induced inhibition of the lysine demethylase KDM4B results in aberrant hypermethylation of histone 3 lysine 9 (H3K9) at loci surrounding DNA breaks, masking a local H3K9 trimethylation signal that is essential for the proper execution of HDR. Consequently, recruitment of TIP60 and ATM, two key proximal HDR factors, is substantially impaired at DNA breaks, with reduced end resection and diminished recruitment of downstream repair factors. These findings provide a mechanistic basis for oncometabolite-induced HDR suppression and may guide effective strategies to exploit these defects for therapeutic gain.


Chromatin/metabolism , DNA Repair , Homologous Recombination , Neoplasms/metabolism , Signal Transduction , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Chromatin/drug effects , DNA Breaks/drug effects , DNA Repair/drug effects , Homologous Recombination/drug effects , Humans , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Lysine Acetyltransferase 5/metabolism , Methylation/drug effects , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Signal Transduction/drug effects
4.
Molecules ; 25(3)2020 Feb 08.
Article En | MEDLINE | ID: mdl-32046275

Unusual nucleic acid structures are salient triggers of endogenous repair and can occur in sequence-specific contexts. Peptide nucleic acids (PNAs) rely on these principles to achieve non-enzymatic gene editing. By forming high-affinity heterotriplex structures within the genome, PNAs have been used to correct multiple human disease-relevant mutations with low off-target effects. Advances in molecular design, chemical modification, and delivery have enabled systemic in vivo application of PNAs resulting in detectable editing in preclinical mouse models. In a model of ß-thalassemia, treated animals demonstrated clinically relevant protein restoration and disease phenotype amelioration, suggesting a potential for curative therapeutic application of PNAs to monogenic disorders. This review discusses the rationale and advances of PNA technologies and their application to gene editing with an emphasis on structural biochemistry and repair.


Cystic Fibrosis/therapy , DNA/genetics , Gene Editing/methods , Genetic Therapy/methods , Peptide Nucleic Acids/genetics , beta-Thalassemia/therapy , Animals , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis/pathology , DNA/metabolism , Disease Models, Animal , Gene Targeting/methods , Gene Transfer Techniques , Humans , Mice , Nanoparticles/chemistry , Nanoparticles/metabolism , Nucleic Acid Conformation , Nucleic Acid Hybridization , Peptide Nucleic Acids/administration & dosage , Peptide Nucleic Acids/metabolism , Recombinational DNA Repair , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , beta-Thalassemia/pathology
...