Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Bio Protoc ; 14(9): e4985, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38737508

ABSTRACT

Pseudouridine (Ψ), the most prevalent modified base in cellular RNAs, has been mapped to numerous sites not only in rRNAs, tRNAs, and snRNAs but also mRNAs. Although there have been multiple techniques to identify Ψs, due to the recent development of sequencing technologies some reagents are not compatible with the current sequencer. Here, we show the updated Pseudo-seq, a technique enabling the genome-wide identification of pseudouridylation sites with single-nucleotide precision. We provide a comprehensive description of Pseudo-seq, covering protocols for RNA isolation from human cells, library preparation, and detailed data analysis procedures. The methodology presented is easily adaptable to any cell or tissue type with high-quality mRNA isolation. It can be used for discovering novel pseudouridylation sites, thus constituting a crucial initial step toward understanding the regulation and function of this modification. Key features • Identification of Ψ sites on mRNAs. • Updated Pseudo-seq provides precise positional and quantitative information of Ψ. • Uses a more efficient library preparation with the latest, currently available materials.

2.
Biochem J ; 481(1): 1-16, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38174858

ABSTRACT

RNA-guided pseudouridylation, a widespread post-transcriptional RNA modification, has recently gained recognition for its role in cellular processes such as pre-mRNA splicing and the modulation of premature termination codon (PTC) readthrough. This review provides insights into its mechanisms, functions, and potential therapeutic applications. It examines the mechanisms governing RNA-guided pseudouridylation, emphasizing the roles of guide RNAs and pseudouridine synthases in catalyzing uridine-to-pseudouridine conversion. A key focus is the impact of RNA-guided pseudouridylation of U2 small nuclear RNA on pre-mRNA splicing, encompassing its influence on branch site recognition and spliceosome assembly. Additionally, the review discusses the emerging role of RNA-guided pseudouridylation in regulating PTC readthrough, impacting translation termination and genetic disorders. Finally, it explores the therapeutic potential of pseudouridine modifications, offering insights into potential treatments for genetic diseases and cancer and the development of mRNA vaccine.


Subject(s)
Pseudouridine , RNA Precursors , Pseudouridine/genetics , Pseudouridine/metabolism , RNA Precursors/metabolism , RNA, Guide, CRISPR-Cas Systems , RNA/metabolism , RNA Processing, Post-Transcriptional , Protein Biosynthesis
3.
Sci Rep ; 13(1): 20420, 2023 11 21.
Article in English | MEDLINE | ID: mdl-37990044

ABSTRACT

Previously, we have identified a novel human metastasis-inducing lncRNA (named SKAI1BC), that suppresses the KAI1/CD82 metastasis-suppressing gene and is upregulated in triple negative breast cancer and melanoma derived cell lines. Modeling of the SKAI1BC lncRNA secondary structure and its potential interaction with Inforna compounds, led us to identify several compounds that might bind the SKAI1BC lncRNA. We found that these compounds inhibit metastasis invasion and cell migration in culture, in all eight types of solid human cancers tested: several of which are the most lethal and/or frequent human malignancies. Moreover, in most cases, the mechanism of action of several of our compounds involves enhancement of KAI1/CD82 RNA level depending on the specific compound and the human tumor type. With the epigenetic inactivation of KAI1/CD82 in at least ten additional solid human cancers, this implies a very good chance to broaden the spectrum of human cancers affected by our compounds. This is the first time that modeling of a large lncRNA (> 700 bp) secondary structure followed by its potential interaction with Inforna like compounds database has led to the identification of potential biologically active small molecule drugs.


Subject(s)
Melanoma , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , Kangai-1 Protein/genetics , Kangai-1 Protein/metabolism , Genes, Tumor Suppressor , Cell Line , Melanoma/drug therapy , Melanoma/genetics , Neoplasm Metastasis
4.
Methods Mol Biol ; 2666: 177-191, 2023.
Article in English | MEDLINE | ID: mdl-37166666

ABSTRACT

Pseudouridine (Ψ) is the most common chemical modification in RNA. In eukaryotes and archaea, pseudouridine synthases, mainly guided by box H/ACA snoRNAs, convert uridine to Ψ. Ψ stabilizes RNA structure and alters RNA-RNA and RNA-protein interactions, conferring important roles in gene expression. Notably, several Ψ-linked human diseases have been identified over the years. In addition, Ψ has also been extensively used in developing mRNA vaccines. Furthermore, it has been shown that pseudouridylation can be site-specifically directed to modify specific nonsense codons, leading to nonsense suppression. All of these, together with a need to better understand the specific functions of Ψs, have motivated the development of in vitro pseudouridylation assays using purified and reconstituted box H/ACA RNPs. Here, we describe an in vitro system for box H/ACA RNA-guided RNA pseudouridylation using human cell extracts. We show that a half guide RNA (only one hairpin) is just as functionally competent as the full-length guide RNA (two hairpins) in guiding site-specific pseudouridylation in the human cell extracts. This discovery offers the opportunity for direct delivery of a short guide RNA to human cells to promote site-specific nonsense suppression and therefore has potential clinical applications.


Subject(s)
Pseudouridine , RNA, Small Nucleolar , Humans , Cell Extracts , Pseudouridine/genetics , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Catalysis
5.
Mol Cell ; 83(4): 637-651.e9, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36764303

ABSTRACT

Nonsense mutations create premature termination codons (PTCs), activating the nonsense-mediated mRNA decay (NMD) pathway to degrade most PTC-containing mRNAs. The undegraded mRNA is translated, but translation terminates at the PTC, leading to no production of the full-length protein. This work presents targeted PTC pseudouridylation, an approach for nonsense suppression in human cells. Specifically, an artificial box H/ACA guide RNA designed to target the mRNA PTC can suppress both NMD and premature translation termination in various sequence contexts. Targeted pseudouridylation exhibits a level of suppression comparable with that of aminoglycoside antibiotic treatments. When targeted pseudouridylation is combined with antibiotic treatment, a much higher level of suppression is observed. Transfection of a disease model cell line (carrying a chromosomal PTC) with a designer guide RNA gene targeting the PTC also leads to nonsense suppression. Thus, targeted pseudouridylation is an RNA-directed gene-specific approach that suppresses NMD and concurrently promotes PTC readthrough.


Subject(s)
Codon, Nonsense , Protein Biosynthesis , Humans , Codon, Nonsense/genetics , Nonsense Mediated mRNA Decay , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Sci Transl Med ; 13(617): eabd5991, 2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34705518

ABSTRACT

The most common cause of amyotrophic lateral sclerosis and frontotemporal dementia (c9ALS/FTD) is an expanded G4C2 RNA repeat [r(G4C2)exp] in chromosome 9 open reading frame 72 (C9orf72), which elicits pathology through several mechanisms. Here, we developed and characterized a small molecule for targeted degradation of r(G4C2)exp. The compound was able to selectively bind r(G4C2)exp's structure and to assemble an endogenous nuclease onto the target, provoking removal of the transcript by native RNA quality control mechanisms. In c9ALS patient­derived spinal neurons, the compound selectively degraded the mutant C9orf72 allele with limited off-targets and reduced quantities of toxic dipeptide repeat proteins (DPRs) translated from r(G4C2)exp. In vivo work in a rodent model showed that abundance of both the mutant allele harboring the repeat expansion and DPRs were selectively reduced by this compound. These results demonstrate that targeted small-molecule degradation of r(G4C2)exp is a strategy for mitigating c9ALS/FTD-associated pathologies and studying disease-associated pathways in preclinical models.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Amyotrophic Lateral Sclerosis/genetics , C9orf72 Protein/genetics , DNA Repeat Expansion , Frontotemporal Dementia/genetics , Humans , Ribonucleases
7.
J Med Chem ; 64(12): 8474-8485, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34101465

ABSTRACT

Myotonic dystrophy type 2 (DM2) is one of >40 microsatellite disorders caused by RNA repeat expansions. The DM2 repeat expansion, r(CCUG)exp (where "exp" denotes expanded repeating nucleotides), is harbored in intron 1 of the CCHC-type zinc finger nucleic acid binding protein (CNBP). The expanded RNA repeat causes disease by a gain-of-function mechanism, sequestering various RNA-binding proteins including the pre-mRNA splicing regulator MBNL1. Sequestration of MBNL1 results in its loss-of-function and concomitant deregulation of the alternative splicing of its native substrates. Notably, this r(CCUG)exp causes retention of intron 1 in the mature CNBP mRNA. Herein, we report druglike small molecules that bind the structure adopted by r(CCUG)exp and improve DM2-associated defects. These small molecules were optimized from screening hits from an RNA-focused small-molecule library to afford a compound that binds r(CCUG)exp specifically and with nanomolar affinity, facilitates endogenous degradation of the aberrantly retained intron in which it is harbored, and rescues alternative splicing defects.


Subject(s)
Benzothiazoles/pharmacology , Quinazolines/pharmacology , RNA/drug effects , Benzothiazoles/chemical synthesis , Humans , Molecular Structure , Myotonic Dystrophy/genetics , Quinazolines/chemical synthesis , RNA/genetics , RNA/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Repetitive Sequences, Nucleic Acid/genetics , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
8.
Cell Chem Biol ; 28(1): 34-45.e6, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33157036

ABSTRACT

Many diseases are caused by toxic RNA repeats. Herein, we designed a lead small molecule that binds the structure of the r(CUG) repeat expansion [r(CUG)exp] that causes myotonic dystrophy type 1 (DM1) and Fuchs endothelial corneal dystrophy (FECD) and rescues disease biology in patient-derived cells and in vivo. Interestingly, the compound's downstream effects are different in the two diseases, owing to the location of the repeat expansion. In DM1, r(CUG)exp is harbored in the 3' untranslated region, and the compound has no effect on the mRNA's abundance. In FECD, however, r(CUG)exp is located in an intron, and the small molecule facilitates excision of the intron, which is then degraded by the RNA exosome complex. Thus, structure-specific, RNA-targeting small molecules can act disease specifically to affect biology, either by disabling the gain-of-function mechanism (DM1) or by stimulating quality control pathways to rid a disease-affected cell of a toxic RNA (FECD).


Subject(s)
Exosomes/drug effects , Fuchs' Endothelial Dystrophy/drug therapy , Myotonic Dystrophy/drug therapy , Small Molecule Libraries/pharmacology , Trinucleotide Repeat Expansion/drug effects , Cells, Cultured , Exosomes/metabolism , Female , Fuchs' Endothelial Dystrophy/metabolism , Humans , Male , Myotonic Dystrophy/metabolism , Trinucleotide Repeat Expansion/genetics
9.
ACS Chem Biol ; 15(12): 3112-3123, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33196168

ABSTRACT

Genetically defined amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), collectively named c9ALS/FTD, are triggered by hexanucleotide GGGGCC repeat expansions [r(G4C2)exp] within the C9orf72 gene. In these diseases, neuronal loss occurs through an interplay of deleterious phenotypes, including r(G4C2)exp RNA gain-of-function mechanisms. Herein, we identified a benzimidazole derivative, CB096, that specifically binds to a repeating 1 × 1 GG internal loop structure, 5'CGG/3'GGC, that is formed when r(G4C2)exp folds. Structure-activity relationship (SAR) studies and molecular dynamics (MD) simulations were used to define the molecular interactions formed between CB096 and r(G4C2)exp that results in the rescue of disease-associated pathways. Overall, this study reveals a unique structural feature within r(G4C2)exp that can be exploited for the development of lead medicines and chemical probes.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Frontotemporal Dementia/genetics , RNA/genetics , Small Molecule Libraries/chemistry , C9orf72 Protein/genetics , G-Quadruplexes , High-Throughput Screening Assays , Humans , Molecular Dynamics Simulation , Molecular Structure , RNA/drug effects , Small Molecule Libraries/pharmacology
10.
ACS Cent Sci ; 6(10): 1713-1721, 2020 Oct 28.
Article in English | MEDLINE | ID: mdl-33140033

ABSTRACT

COVID-19 is a global pandemic, thus requiring multiple strategies to develop modalities against it. Herein, we designed multiple bioactive small molecules that target a functional structure within the SARS-CoV-2's RNA genome, the causative agent of COVID-19. An analysis to characterize the structure of the RNA genome provided a revised model of the SARS-CoV-2 frameshifting element, in particular its attenuator hairpin. By studying an RNA-focused small molecule collection, we identified a drug-like small molecule (C5) that avidly binds to the revised attenuator hairpin structure with a K d of 11 nM. The compound stabilizes the hairpin's folded state and impairs frameshifting in cells. The ligand was further elaborated into a ribonuclease targeting chimera (RIBOTAC) to recruit a cellular ribonuclease to destroy the viral genome (C5-RIBOTAC) and into a covalent molecule (C5-Chem-CLIP) that validated direct target engagement and demonstrated its specificity for the viral RNA, as compared to highly expressed host mRNAs. The RIBOTAC lead optimization strategy improved the bioactivity of the compound at least 10-fold. Collectively, these studies demonstrate that the SARS-CoV-2 RNA genome should be considered druggable.

11.
J Am Chem Soc ; 142(19): 8706-8727, 2020 05 13.
Article in English | MEDLINE | ID: mdl-32364710

ABSTRACT

Approximately 95% of human genes are alternatively spliced, and aberrant splicing events can cause disease. One pre-mRNA that is alternatively spliced and linked to neurodegenerative diseases is tau (microtubule-associated protein tau), which can cause frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) and can contribute to Alzheimer's disease. Here, we describe the design of structure-specific lead small molecules that directly target tau pre-mRNA from sequence. This was followed by hit expansion and analogue synthesis to further improve upon these initial lead molecules. The emergent compounds were assessed for functional activity in a battery of assays, including binding assays and an assay that mimics molecular recognition of tau pre-mRNA by a U1 small nuclear ribonucleoprotein (snRNP) splicing factor. Compounds that emerged from these studies had enhanced potency and selectivity for the target RNA relative to the initial hits, while also having significantly improved drug-like properties. The compounds are shown to directly target tau pre-mRNA in cells, via chemical cross-linking and isolation by pull-down target profiling, and to rescue disease-relevant splicing of tau pre-mRNA in a variety of cellular systems, including primary neurons. More broadly, this study shows that lead, structure-specific compounds can be designed from sequence and then further optimized for their physicochemical properties while at the same time enhancing their activity.


Subject(s)
RNA Splicing/drug effects , RNA, Messenger/antagonists & inhibitors , Small Molecule Libraries/pharmacology , tau Proteins/antagonists & inhibitors , HeLa Cells , Humans , Models, Molecular , Molecular Structure , RNA Splicing/genetics , RNA, Messenger/genetics , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Thermodynamics , tau Proteins/genetics
12.
SLAS Discov ; 25(8): 869-894, 2020 09.
Article in English | MEDLINE | ID: mdl-32419578

ABSTRACT

RNA molecules have a variety of cellular functions that can drive disease pathologies. They are without a doubt one of the most intriguing yet controversial small-molecule drug targets. The ability to widely target RNA with small molecules could be revolutionary, once the right tools, assays, and targets are selected, thereby defining which biomolecules are targetable and what constitutes drug-like small molecules. Indeed, approaches developed over the past 5-10 years have changed the face of small molecule-RNA targeting by addressing historic concerns regarding affinity, selectivity, and structural dynamics. Presently, selective RNA-protein complex stabilizing drugs such as branaplam and risdiplam are in clinical trials for the modulation of SMN2 splicing, compounds identified from phenotypic screens with serendipitous outcomes. Fully developing RNA as a druggable target will require a target engagement-driven approach, and evolving chemical collections will be important for the industrial development of this class of target. In this review we discuss target-directed approaches that can be used to identify RNA-binding compounds and the chemical knowledge we have today of small-molecule RNA binders.


Subject(s)
Molecular Targeted Therapy , RNA/drug effects , Small Molecule Libraries/chemistry , Azo Compounds/therapeutic use , Drug Design , Humans , Multiprotein Complexes/genetics , Neuromuscular Agents/therapeutic use , Pyrimidines/therapeutic use , RNA/genetics , RNA Splicing/drug effects , RNA Splicing/genetics , Small Molecule Libraries/therapeutic use , Survival of Motor Neuron 2 Protein/genetics
13.
Ann N Y Acad Sci ; 1471(1): 57-71, 2020 07.
Article in English | MEDLINE | ID: mdl-30964958

ABSTRACT

Aberrant RNA structure and function operate in neurological disease progression and severity. As RNA contributes to disease pathology in a complex fashion, that is, via various mechanisms, it has become an attractive therapeutic target for small molecules and oligonucleotides. In this review, we discuss the identification of RNA structures that cause or contribute to neurological diseases as well as recent progress toward the development of small molecules that target them, including small molecule modulators of pre-mRNA splicing and RNA repeat expansions that cause microsatellite disorders such as Huntington's disease and amyotrophic lateral sclerosis. The use of oligonucleotide-based modalities is also discussed. There are key differences between small molecule and oligonucleotide targeting of RNA. The former targets RNA structure, while the latter prefers unstructured regions. Thus, some targets will be preferentially targeted by oligonucleotides and others by small molecules.


Subject(s)
Nervous System Diseases/drug therapy , Nucleic Acid Conformation/drug effects , RNA/genetics , Small Molecule Libraries/therapeutic use , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Humans , Huntington Disease/drug therapy , Huntington Disease/genetics , Nervous System Diseases/genetics , Nervous System Diseases/pathology , Oligonucleotides/genetics , Oligonucleotides/therapeutic use , RNA/antagonists & inhibitors , RNA Precursors/genetics , RNA Splicing/drug effects
14.
PLoS One ; 14(7): e0219210, 2019.
Article in English | MEDLINE | ID: mdl-31291322

ABSTRACT

Tauopathies are neurodegenerative diseases that affect millions of people worldwide including those with Alzheimer's disease. While many efforts have focused on understanding the role of tau protein in neurodegeneration, there has been little done to systematically analyze and study the structures within tau's encoding RNA and their connection to disease pathology. Knowledge of RNA structure can provide insights into disease mechanisms and how to affect protein production for therapeutic benefit. Using computational methods based on thermodynamic stability and evolutionary conservation, we identified structures throughout the tau pre-mRNA, especially at exon-intron junctions and within the 5' and 3' untranslated regions (UTRs). In particular, structures were identified at twenty exon-intron junctions. The 5' UTR contains one structured region, which lies within a known internal ribosome entry site. The 3' UTR contains eight structured regions, including one that contains a polyadenylation signal. A series of functional experiments were carried out to assess the effects of mutations associated with mis-regulation of alternative splicing of exon 10 and to identify regions of the 3' UTR that contain cis-regulatory elements. These studies defined novel structural regions within the mRNA that affect stability and pre-mRNA splicing and may lead to new therapeutic targets for treating tau-associated diseases.


Subject(s)
RNA Precursors/chemistry , RNA, Messenger/genetics , Tauopathies/genetics , tau Proteins/genetics , 3' Untranslated Regions/genetics , Alternative Splicing/genetics , Alzheimer Disease , Exons/genetics , Humans , Introns/genetics , Mutation , Nucleic Acid Conformation , Polyadenylation/genetics , RNA Precursors/genetics , RNA, Messenger/chemistry , Tauopathies/pathology , tau Proteins/chemistry
15.
PLoS One ; 14(6): e0213758, 2019.
Article in English | MEDLINE | ID: mdl-31206539

ABSTRACT

The MYC gene encodes a human transcription factor and proto-oncogene that is dysregulated in over half of all known cancers. To better understand potential post-transcriptional regulatory features affecting MYC expression, we analyzed secondary structures in the MYC mRNA using a program that is optimized for finding small locally-folded motifs with a high propensity for function. This was accomplished by calculating folding metrics across the MYC sequence using a sliding analysis window and generating unique consensus base pairing models weighted by their lower-than-random predicted folding energy. A series of 30 motifs were identified, primarily in the 5' and 3' untranslated regions, which show evidence of structural conservation and compensating mutations across vertebrate MYC homologs. This analysis was able to recapitulate known elements found within an internal ribosomal entry site, as well as discover a novel element in the 3' UTR that is unusually stable and conserved. This novel motif was shown to affect MYC expression, potentially via the modulation of miRNA target accessibility or other trans-regulatory factors. In addition to providing basic insights into mechanisms that regulate MYC expression, this study provides numerous, potentially druggable RNA targets for the MYC gene, which is considered "undruggable" at the protein level.


Subject(s)
Conserved Sequence , Gene Expression , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/genetics , 3' Untranslated Regions , 5' Untranslated Regions , Gene Expression Regulation , Humans , MicroRNAs/genetics , Proto-Oncogene Mas
16.
Proc Natl Acad Sci U S A ; 116(16): 7799-7804, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30926669

ABSTRACT

Myotonic dystrophy type 1 (DM1) is an incurable neuromuscular disorder caused by an expanded CTG repeat that is transcribed into r(CUG)exp The RNA repeat expansion sequesters regulatory proteins such as Muscleblind-like protein 1 (MBNL1), which causes pre-mRNA splicing defects. The disease-causing r(CUG)exp has been targeted by antisense oligonucleotides, CRISPR-based approaches, and RNA-targeting small molecules. Herein, we describe a designer small molecule, Cugamycin, that recognizes the structure of r(CUG)exp and cleaves it in both DM1 patient-derived myotubes and a DM1 mouse model, leaving short repeats of r(CUG) untouched. In contrast, oligonucleotides that recognize r(CUG) sequence rather than structure cleave both long and short r(CUG)-containing transcripts. Transcriptomic, histological, and phenotypic studies demonstrate that Cugamycin broadly and specifically relieves DM1-associated defects in vivo without detectable off-targets. Thus, small molecules that bind and cleave RNA have utility as lead chemical probes and medicines and can selectively target disease-causing RNA structures to broadly improve defects in preclinical animal models.


Subject(s)
Bleomycin/analogs & derivatives , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Oligonucleotides/chemistry , RNA Splicing/genetics , RNA/genetics , RNA/metabolism , Trinucleotide Repeat Expansion/genetics , Animals , Bleomycin/chemistry , Disease Models, Animal , Drug Design , Humans , Mice , Oligonucleotides/metabolism
17.
Nucleic Acids Res ; 46(10): 4883-4892, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29718397

ABSTRACT

RNA folding free energy change nearest neighbor parameters are widely used to predict folding stabilities of secondary structures. They were determined by linear regression to datasets of optical melting experiments on small model systems. Traditionally, the optical melting experiments are analyzed assuming a two-state model, i.e. a structure is either complete or denatured. Experimental evidence, however, shows that structures exist in an ensemble of conformations. Partition functions calculated with existing nearest neighbor parameters predict that secondary structures can be partially denatured, which also directly conflicts with the two-state model. Here, a new approach for determining RNA nearest neighbor parameters is presented. Available optical melting data for 34 Watson-Crick helices were fit directly to a partition function model that allows an ensemble of conformations. Fitting parameters were the enthalpy and entropy changes for helix initiation, terminal AU pairs, stacks of Watson-Crick pairs and disordered internal loops. The resulting set of nearest neighbor parameters shows a 38.5% improvement in the sum of residuals in fitting the experimental melting curves compared to the current literature set.


Subject(s)
Computational Biology/methods , Models, Chemical , RNA/chemistry , Algorithms , Entropy , Nucleic Acid Conformation , Thermodynamics , Transition Temperature
18.
Chem Rev ; 118(4): 1599-1663, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29322778

ABSTRACT

Rapid progress in genome sequencing technology has put us firmly into a postgenomic era. A key challenge in biomedical research is harnessing genome sequence to fulfill the promise of personalized medicine. This Review describes how genome sequencing has enabled the identification of disease-causing biomolecules and how these data have been converted into chemical probes of function, preclinical lead modalities, and ultimately U.S. Food and Drug Administration (FDA)-approved drugs. In particular, we focus on the use of oligonucleotide-based modalities to target disease-causing RNAs; small molecules that target DNA, RNA, or protein; the rational repurposing of known therapeutic modalities; and the advantages of pharmacogenetics. Lastly, we discuss the remaining challenges and opportunities in the direct utilization of genome sequence to enable design of medicines.


Subject(s)
Genome, Human , Molecular Probes/chemistry , Cell Line, Tumor , Drug Repositioning , High-Throughput Nucleotide Sequencing , Humans , Oligonucleotides/pharmacology , Oligonucleotides/therapeutic use , Pharmacogenetics , Proteins/drug effects , RNA/chemistry , Small Molecule Libraries , United States , United States Food and Drug Administration
19.
Biochemistry ; 56(27): 3463-3474, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28617590

ABSTRACT

RNA repeat expansions cause a host of incurable, genetically defined diseases. The most common class of RNA repeats consists of trinucleotide repeats. These long, repeating transcripts fold into hairpins containing 1 × 1 internal loops that can mediate disease via a variety of mechanism(s) in which RNA is the central player. Two of these disorders are Huntington's disease and myotonic dystrophy type 1, which are caused by r(CAG) and r(CUG) repeats, respectively. We report the structures of two RNA constructs containing three copies of a r(CAG) [r(3×CAG)] or r(CUG) [r(3×CUG)] motif that were modeled with nuclear magnetic resonance spectroscopy and simulated annealing with restrained molecular dynamics. The 1 × 1 internal loops of r(3×CAG) are stabilized by one-hydrogen bond (cis Watson-Crick/Watson-Crick) AA pairs, while those of r(3×CUG) prefer one- or two-hydrogen bond (cis Watson-Crick/Watson-Crick) UU pairs. Assigned chemical shifts for the residues depended on the identity of neighbors or next nearest neighbors. Additional insights into the dynamics of these RNA constructs were gained by molecular dynamics simulations and a discrete path sampling method. Results indicate that the global structures of the RNA are A-form and that the loop regions are dynamic. The results will be useful for understanding the dynamic trajectory of these RNA repeats but also may aid in the development of therapeutics.


Subject(s)
Huntingtin Protein/genetics , Huntington Disease/genetics , Models, Molecular , Myotonic Dystrophy/genetics , Myotonin-Protein Kinase/genetics , RNA, Messenger/chemistry , Trinucleotide Repeat Expansion , 3' Untranslated Regions , Base Pairing , Energy Transfer , Exons , Humans , Huntingtin Protein/chemistry , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Hydrogen Bonding , Molecular Dynamics Simulation , Mutation , Myotonic Dystrophy/metabolism , Myotonin-Protein Kinase/chemistry , Myotonin-Protein Kinase/metabolism , Nuclear Magnetic Resonance, Biomolecular , Nucleic Acid Conformation , Nucleotide Motifs , RNA Folding , RNA, Messenger/metabolism , Uridine/analogs & derivatives , Uridine/chemistry
20.
Methods Mol Biol ; 1490: 177-86, 2016.
Article in English | MEDLINE | ID: mdl-27665599

ABSTRACT

Dynamic programming methods for predicting RNA secondary structure often use thermodynamics and experimental restraints and/or constraints to limit folding space. Chemical mapping results typically restrain certain nucleotides not to be in AU or GC pairs. Two-dimensional nuclear magnetic resonance (NMR) spectra can reveal the order of AU, GC, and GU pairs in double helixes. This chapter describes a program, NMR-assisted prediction of secondary structure and chemical shifts (NAPSS-CS), that constrains possible secondary structures on the basis of the NMR determined order and 5'-3' direction of AU, GC, and GU pairs in helixes. NAPSS-CS minimally requires input of the order of base pairs as determined from nuclear Overhauser effect spectroscopy (NOESY) of imino protons. The program deduces the 5'-3' direction of the base pairs if certain chemical shifts are also input. Secondary structures predicted by the program provide assignments of input chemical shifts to particular nucleotides in the sequence, thus facilitating an important step for determination of the three dimensional structure by NMR. The method is particularly useful for revealing pseudoknots and an example is provided. The method may also allow determination of secondary structures when a sequence folds into two structures that exchange slowly.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular , Nucleic Acid Conformation , RNA/chemistry , Animals , Computational Biology/methods , Humans , Web Browser
SELECTION OF CITATIONS
SEARCH DETAIL
...