Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(48): e2210532119, 2022 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-36409902

RESUMO

A hexanucleotide repeat expansion in intron 1 of the C9orf72 gene is the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia, or c9ALS/FTD. The RNA transcribed from the expansion, r(G4C2)exp, causes various pathologies, including intron retention, aberrant translation that produces toxic dipeptide repeat proteins (DPRs), and sequestration of RNA-binding proteins (RBPs) in RNA foci. Here, we describe a small molecule that potently and selectively interacts with r(G4C2)exp and mitigates disease pathologies in spinal neurons differentiated from c9ALS patient-derived induced pluripotent stem cells (iPSCs) and in two c9ALS/FTD mouse models. These studies reveal a mode of action whereby a small molecule diminishes intron retention caused by the r(G4C2)exp and allows the liberated intron to be eliminated by the nuclear RNA exosome, a multi-subunit degradation complex. Our findings highlight the complexity of mechanisms available to RNA-binding small molecules to alleviate disease pathologies and establishes a pipeline for the design of brain penetrant small molecules targeting RNA with novel modes of action in vivo.


Assuntos
Exossomos , Demência Frontotemporal , Animais , Camundongos , Demência Frontotemporal/metabolismo , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , RNA/genética , Exossomos/metabolismo , Barreira Hematoencefálica/metabolismo , Complexo Multienzimático de Ribonucleases do Exossomo/metabolismo , Encéfalo/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , RNA Nuclear
2.
J Med Chem ; 64(12): 8474-8485, 2021 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-34101465

RESUMO

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.


Assuntos
Benzotiazóis/farmacologia , Quinazolinas/farmacologia , RNA/efeitos dos fármacos , Benzotiazóis/síntese química , Humanos , Estrutura Molecular , Distrofia Miotônica/genética , Quinazolinas/síntese química , RNA/genética , RNA/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Sequências Repetitivas de Ácido Nucleico/genética , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/farmacologia , Relação Estrutura-Atividade
3.
ACS Med Chem Lett ; 12(6): 907-914, 2021 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-34141068

RESUMO

RNA contributes to disease pathobiology and is an important therapeutic target. The downstream biology of disease-causing RNAs can be short-circuited with small molecules that recognize structured regions. The discovery and optimization of small molecules interacting with RNA is, however, challenging. Herein, we demonstrate a massively parallel one-bead-one-compound methodology, employed to optimize the linker region of a dimeric compound that binds the toxic r(CUG) repeat expansion [r(CUG)exp] causative of myotonic dystrophy type 1 (DM1). Indeed, affinity selection on a 331,776-member library allowed the discovery of a compound with enhanced potency both in vitro (10-fold) and in DM1-patient-derived myotubes (5-fold). Molecular dynamics simulations revealed additional interactions between the optimized linker and the RNA, resulting in ca. 10 kcal/mol lower binding free energy. The compound was conjugated to a cleavage module, which directly cleaved the transcript harboring the r(CUG)exp and alleviated disease-associated defects.

4.
Cell Chem Biol ; 28(1): 34-45.e6, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33157036

RESUMO

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).


Assuntos
Exossomos/efeitos dos fármacos , Distrofia Endotelial de Fuchs/tratamento farmacológico , Distrofia Miotônica/tratamento farmacológico , Bibliotecas de Moléculas Pequenas/farmacologia , Expansão das Repetições de Trinucleotídeos/efeitos dos fármacos , Células Cultivadas , Exossomos/metabolismo , Feminino , Distrofia Endotelial de Fuchs/metabolismo , Humanos , Masculino , Distrofia Miotônica/metabolismo , Expansão das Repetições de Trinucleotídeos/genética
5.
Chem Soc Rev ; 49(20): 7252-7270, 2020 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-32935689

RESUMO

The design and discovery of small molecule medicines has largely been focused on a small number of druggable protein families. A new paradigm is emerging, however, in which small molecules exert a biological effect by interacting with RNA, both to study human disease biology and provide lead therapeutic modalities. Due to this potential for expanding target pipelines and treating a larger number of human diseases, robust platforms for the rational design and optimization of small molecules interacting with RNAs (SMIRNAs) are in high demand. This review highlights three major pillars in this area. First, the transcriptome-wide identification and validation of structured RNA elements, or motifs, within disease-causing RNAs directly from sequence is presented. Second, we provide an overview of high-throughput screening approaches to identify SMIRNAs as well as discuss the lead identification strategy, Inforna, which decodes the three-dimensional (3D) conformation of RNA motifs with small molecule binding partners, directly from sequence. An emphasis is placed on target validation methods to study the causality between modulating the RNA motif in vitro and the phenotypic outcome in cells. Third, emergent modalities that convert occupancy-driven mode of action SMIRNAs into event-driven small molecule chemical probes, such as RNA cleavers and degraders, are presented. Finally, the future of the small molecule RNA therapeutics field is discussed, as well as hurdles to overcome to develop potent and selective RNA-centric chemical probes.


Assuntos
RNA/química , Bibliotecas de Moléculas Pequenas/química , Antagomirs/química , Antagomirs/metabolismo , Desenho de Fármacos , Doença de Huntington/genética , Doença de Huntington/patologia , MicroRNAs/química , MicroRNAs/metabolismo , Conformação de Ácido Nucleico , RNA/metabolismo , RNA Viral/química , RNA Viral/metabolismo
6.
J Med Chem ; 63(14): 7827-7839, 2020 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-32657583

RESUMO

RNA repeat expansions are responsible for more than 30 incurable diseases. Among them is myotonic dystrophy type 1 (DM1), the most common form of adult on-set muscular dystrophy. DM1 is caused by an r(CUG) repeat expansion [r(CUG)exp] located in the 3' untranslated region (UTR) of the dystrophia myotonica protein kinase gene. This repeat expansion is highly structured, forming a periodic array of 5'CUG/3'GUC internal loop motifs. We therefore designed dimeric compounds that simultaneously bind two of these motifs by connecting two RNA-binding modules with peptoid linkers of different geometries and lengths. The optimal linker contains two proline residues and enhances compound affinity. Equipping this molecule with a bleomycin A5 cleaving module converts the simple binding compound into a potent allele-selective cleaver of r(CUG)exp. This study shows that the linker in modularly assembled ligands targeting RNA can be optimized to afford potent biological activity.


Assuntos
Benzimidazóis/farmacologia , Oligopeptídeos/farmacologia , RNA/química , Benzimidazóis/síntese química , Bleomicina/análogos & derivados , Bleomicina/síntese química , Bleomicina/farmacologia , Dano ao DNA/efeitos dos fármacos , Desenho de Fármacos , Humanos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Oligopeptídeos/síntese química , RNA/efeitos dos fármacos , RNA/genética , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/farmacologia , Sequências de Repetição em Tandem/efeitos dos fármacos
7.
ACS Chem Biol ; 15(7): 1820-1825, 2020 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-32551539

RESUMO

Expanded RNA repeats cause more than 30 incurable diseases. One approach to mitigate their toxicity is by using small molecules that assemble into potent, oligomeric species upon binding to the disease-causing RNA in cells. Herein, we show that the expanded repeat [r(CUG)exp] that causes myotonic dystrophy type 1 (DM1) catalyzes the in situ synthesis of its own inhibitor using an RNA-templated tetrazine ligation in DM1 patient-derived cells. The compound synthesized on-site improved DM1-associated defects at picomolar concentrations, enhancing potency by 10 000-fold, compared to its parent compounds that cannot undergo oligomerization. A fluorogenic reaction is also described where r(CUG)exp templates the synthesis of its own imaging probe to enable visualization of the repeat in its native context in live cells and muscle tissue.


Assuntos
Fluoresceínas/farmacologia , Corantes Fluorescentes/farmacologia , Compostos Heterocíclicos com 1 Anel/farmacologia , Miotonina Proteína Quinase/genética , RNA/antagonistas & inibidores , Animais , Química Click , Fluoresceínas/síntese química , Corantes Fluorescentes/síntese química , Compostos Heterocíclicos com 1 Anel/síntese química , Humanos , Camundongos , Distrofia Miotônica/enzimologia , Distrofia Miotônica/genética , RNA/genética , Sequências de Repetição em Tandem , Transcrição Gênica/efeitos dos fármacos
8.
ACS Chem Biol ; 15(8): 2031-2040, 2020 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-32568503

RESUMO

Selectivity is a key requirement of high-quality chemical probes and lead medicines; however, methods to quantify and compare the selectivity of small molecules have not been standardized across the field. Herein, we discuss the origins and use of a comprehensive, single value term to quantify selectivity, the Gini coefficient. Case studies presented include compounds that target protein kinases, small molecules that bind RNA structures, and small molecule chimeras that bind to and degrade the target RNA. With an increasing number of transcriptome- and proteome-wide studies, we submit that reporting Gini coefficients as a quantitative descriptor of selectivity should be used broadly.


Assuntos
Sondas Moleculares/química , Bibliotecas de Moléculas Pequenas/química , Modelos Químicos , RNA/química
9.
ACS Chem Biol ; 15(4): 849-855, 2020 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-32186845

RESUMO

RNA repeat expansions cause more than 30 neurological and neuromuscular diseases with no known cures. Since repeat expansions operate via diverse pathomechanisms, one potential therapeutic strategy is to rid them from disease-affected cells, using bifunctional small molecules that cleave the aberrant RNA. Such an approach has been previously implemented for the RNA repeat that causes myotonic dystrophy type 1 [DM1, r(CUG)exp] with Cugamycin, which is a small molecule that selectively binds r(CUG)exp conjugated to a bleomycin A5 cleaving module. Herein, we demonstrate that, by replacing bleomycin A5 with deglycobleomycin, an analogue in which the carbohydrate domain of bleomycin A5 is removed, the selectivity of the resulting small-molecule conjugate (DeglycoCugamycin) was enhanced, while maintaining potent and allele-selective cleavage of r(CUG)exp and rescue of DM1-associated defects. In particular, DeglycoCugamycin did not induce the DNA damage that is observed with high concentrations (25 µM) of Cugamycin, while selectively cleaving the disease-causing allele and improving DM1 defects at 1 µM.


Assuntos
Bleomicina/análogos & derivados , Clivagem do DNA/efeitos dos fármacos , DNA/química , Expansão das Repetições de Trinucleotídeos/efeitos dos fármacos , Animais , Bleomicina/química , Bleomicina/farmacologia , Linhagem Celular , Dano ao DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Camundongos , Proteínas de Ligação a RNA/genética
10.
ACS Chem Biol ; 15(2): 485-493, 2020 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-31927948

RESUMO

Myotonic dystrophy type 2 (DM2) is a genetically defined muscular dystrophy that is caused by an expanded repeat of r(CCUG) [r(CCUG)exp] in intron 1 of a CHC-type zinc finger nucleic acid binding protein (CNBP) pre-mRNA. Various mechanisms contribute to DM2 pathology including pre-mRNA splicing defects caused by sequestration of the RNA splicing regulator muscleblind-like-1 (MBNL1) by r(CCUG)exp. Herein, we study the biological impacts of the molecular recognition of r(CCUG)exp's structure by a designer dimeric small molecule that directly cleaves the RNA in patient-derived cells. The compound is comprised of two RNA-binding modules conjugated to a derivative of the natural product bleomycin. Careful design of the chimera affords RNA-specific cleavage, as attachment of the bleomycin cleaving module was done in a manner that disables DNA cleavage. The chimeric cleaver is more potent than the parent binding compound for alleviating DM2-associated defects. Importantly, oligonucleotides targeting the r(CCUG)exp sequence for cleavage exacerbate DM2 defects due to recognition of a short r(CCUG) sequence that is embedded in CNBP, argonaute-1 (AGO1), and MBNL1, reducing their levels. The latter event causes a greater depletion of functional MBNL1 than the amount already sequestered by r(CCUG)exp. Thus, compounds targeting RNA structures can have functional advantages over oligonucleotides that target the sequence in some disease settings, particularly in DM2.


Assuntos
Bleomicina/farmacologia , Clivagem do RNA/efeitos dos fármacos , Proteínas de Ligação a RNA/metabolismo , RNA/efeitos dos fármacos , Sequências Repetitivas de Ácido Nucleico/efeitos dos fármacos , Sequência de Bases , Bleomicina/química , Linhagem Celular , Fibroblastos/efeitos dos fármacos , Humanos , Mutação , Distrofia Miotônica/genética , Proteínas de Ligação a RNA/genética
11.
Cell Chem Biol ; 27(2): 223-231.e4, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-31981476

RESUMO

Myotonic dystrophy type 2 (DM2) is a genetically defined disease caused by a toxic expanded repeat of r(CCUG) [r(CCUG)exp], harbored in intron 1 of CCHC-type zinc-finger nucleic acid binding protein (CNBP) pre-mRNA. This r(CCUG)exp causes toxicity via a gain-of-function mechanism, resulting in three pathological hallmarks: aggregation into nuclear foci; sequestration of muscleblind-like-1 (MBNL1) protein, leading to splicing defects; and retention of CNBP intron 1. We studied two types of small molecules with different modes of action, ones that simply bind and ones that are templated by r(CCUG)exp in cells, i.e., the RNA synthesizes its own drug. Indeed, our studies completed in DM2 patient-derived fibroblasts showed that the compounds disrupt the r(CCUG)exp-MBNL1 complex, reduce intron retention, subjecting the liberated intronic r(CCUG)exp to native decay pathways, and rescue other DM2-associated cellular defects. Importantly, this study shows that small molecules can modulate RNA biology by shunting toxic transcripts toward native decay pathways.


Assuntos
Distrofia Miotônica/patologia , Proteínas de Ligação a RNA/genética , RNA/metabolismo , Sequências de Repetição em Tandem/genética , Processamento Alternativo/efeitos dos fármacos , Linhagem Celular , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Íntrons , Distrofia Miotônica/genética , Interferência de RNA , Precursores de RNA/metabolismo , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/metabolismo , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia
12.
Ann N Y Acad Sci ; 1471(1): 57-71, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-30964958

RESUMO

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.


Assuntos
Doenças do Sistema Nervoso/tratamento farmacológico , Conformação de Ácido Nucleico/efeitos dos fármacos , RNA/genética , Bibliotecas de Moléculas Pequenas/uso terapêutico , Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/genética , Humanos , Doença de Huntington/tratamento farmacológico , Doença de Huntington/genética , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/patologia , Oligonucleotídeos/genética , Oligonucleotídeos/uso terapêutico , RNA/antagonistas & inibidores , Precursores de RNA/genética , Splicing de RNA/efeitos dos fármacos
13.
Proc Natl Acad Sci U S A ; 116(16): 7799-7804, 2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-30926669

RESUMO

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.


Assuntos
Bleomicina/análogos & derivados , Distrofia Miotônica/genética , Distrofia Miotônica/metabolismo , Oligonucleotídeos/química , Splicing de RNA/genética , RNA/genética , RNA/metabolismo , Expansão das Repetições de Trinucleotídeos/genética , Animais , Bleomicina/química , Modelos Animais de Doenças , Desenho de Fármacos , Humanos , Camundongos , Oligonucleotídeos/metabolismo
14.
Cell Chem Biol ; 25(9): 1086-1094.e7, 2018 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-30251629

RESUMO

Potential RNA drug targets for small molecules are found throughout the human transcriptome, yet small molecules known to elicit a pharmacological response by directly targeting RNA are limited to antibacterials. Herein, we describe AbsorbArray, a small molecule microarray-based approach that allows for unmodified compounds, including FDA-approved drugs, to be probed for binding to RNA motif libraries in a massively parallel format. Several drug classes bind RNA including kinase and topoisomerase inhibitors. The latter avidly bound the motif found in the Dicer site of oncogenic microRNA (miR)-21 and inhibited its processing both in vitro and in cells. The most potent compound de-repressed a downstream protein target and inhibited a miR-21-mediated invasive phenotype. The compound's activity was ablated upon overexpression of pre-miR-21. Target validation via chemical crosslinking and isolation by pull-down showed direct engagement of pre-miR-21 by the small molecule in cells, demonstrating that RNAs should indeed be considered druggable.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Desenho de Fármacos , MicroRNAs/metabolismo , Neoplasias/tratamento farmacológico , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Linhagem Celular Tumoral , Aprovação de Drogas , Descoberta de Drogas/métodos , Humanos , MicroRNAs/genética , Terapia de Alvo Molecular , Neoplasias/genética , Neoplasias/metabolismo
15.
Chem Rev ; 118(4): 1599-1663, 2018 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-29322778

RESUMO

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.


Assuntos
Genoma Humano , Sondas Moleculares/química , Linhagem Celular Tumoral , Reposicionamento de Medicamentos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Oligonucleotídeos/farmacologia , Oligonucleotídeos/uso terapêutico , Farmacogenética , Proteínas/efeitos dos fármacos , RNA/química , Bibliotecas de Moléculas Pequenas , Estados Unidos , United States Food and Drug Administration
16.
Chembiochem ; 19(1): 43-47, 2018 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-29084369

RESUMO

Noncoding RNAs are pervasive in cells and contribute to diseases such as cancer. A question in biomedical research is whether noncoding RNAs are targets of medicines. Bleomycin is a natural product that cleaves DNA; however, it is known to cleave RNA in vitro. Herein, an in-depth analysis of the RNA cleavage preferences of bleomycin A5 is presented. Bleomycin A5 prefers to cleave RNAs with stretches of AU base pairs. Based on these preferences and bioinformatic analysis, the microRNA-10b hairpin precursor was identified as a potential substrate for bleomycin A5. Both in vitro and cellular experiments demonstrated cleavage. Importantly, chemical cleavage by bleomycin A5 in the microRNA-10b hairpin precursors occurred near the Drosha and Dicer enzymatic processing sites and led to destruction of the microRNA. Evidently, oncogenic noncoding RNAs can be considered targets of cancer medicines and might elicit their pharmacological effects by targeting noncoding RNA.


Assuntos
Bleomicina/metabolismo , RNA não Traduzido/metabolismo , Sequência de Bases , Bleomicina/química , Células HeLa , Humanos , MicroRNAs/química , MicroRNAs/metabolismo , Conformação de Ácido Nucleico , Clivagem do RNA , RNA não Traduzido/química , Ribonuclease III/metabolismo
17.
Acc Chem Res ; 49(12): 2698-2704, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-27993012

RESUMO

The discovery of RNA catalysis in the 1980s and the dissemination of the human genome sequence at the start of this century inspired investigations of the regulatory roles of noncoding RNAs in biology. In fact, the Encyclopedia of DNA Elements (ENCODE) project has shown that only 1-2% of the human genome encodes protein, yet 75% is transcribed into RNA. Functional studies both preceding and following the ENCODE project have shown that these noncoding RNAs have important roles in regulating gene expression, developmental timing, and other critical functions. RNA's diverse roles are often a consequence of the various folds that it adopts. The single-stranded nature of the biopolymer enables it to adopt intramolecular folds with noncanonical pairings to lower its free energy. These folds can be scaffolds to bind proteins or to form frameworks to interact with other RNAs. Not surprisingly, dysregulation of certain noncoding RNAs has been shown to be causative of disease. Given this as the background, it is easy to see why it would be useful to develop methods that target RNA and manipulate its biology in rational and predictable ways. The antisense approach has afforded strategies to target RNAs via Watson-Crick base pairing and has typically focused on targeting partially unstructured regions of RNA. Small molecule strategies to target RNA would be desirable not only because compounds could be lead optimized via medicinal chemistry but also because structured regions within an RNA of interest could be targeted to directly interfere with RNA folds that contribute to disease. Additionally, small molecules have historically been the most successful drug candidates. Until recently, the ability to design small molecules that target non-ribosomal RNAs has been elusive, creating the perception that they are "undruggable". In this Account, approaches to demystify targeting RNA with small molecules are described. Rather than bulk screening for compounds that bind to singular targets, which is the purview of the pharmaceutical industry and academic institutions with high throughput screening facilities, we focus on methods that allow for the rational design of small molecules toward biological RNAs. One enabling and foundational technology that has been developed is two-dimensional combinatorial screening (2DCS), a library-versus-library selection approach that allows the identification of the RNA motif binding preferences of small molecules from millions of combinations. A landscape map of the 2DCS-defined and annotated RNA motif-small molecule interactions is then placed into Inforna, a computational tool that allows one to mine these interactions against an RNA of interest or an entire transcriptome. Indeed, this approach has been enabled by tools to annotate RNA structure from sequence, an invaluable asset to the RNA community and this work, and has allowed for the rational identification of "druggable" RNAs in a target agnostic fashion.


Assuntos
Antineoplásicos/farmacologia , MicroRNAs/antagonistas & inibidores , Animais , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Benzimidazóis/química , Benzimidazóis/metabolismo , Benzimidazóis/farmacocinética , Benzimidazóis/farmacologia , Linhagem Celular Tumoral , Técnicas de Química Combinatória , Desenho de Fármacos , Proteína Forkhead Box O1/metabolismo , Humanos , MicroRNAs/química , Análise em Microsséries , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Relação Estrutura-Atividade
18.
Bioorg Med Chem Lett ; 26(23): 5792-5796, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27839685

RESUMO

RNA is an important drug target, but current approaches to identify bioactive small molecules have been engineered primarily for protein targets. Moreover, the identification of small molecules that bind a specific RNA target with sufficient potency remains a challenge. Computer-aided drug design (CADD) and, in particular, ligand-based drug design provide a myriad of tools to identify rapidly new chemical entities for modulating a target based on previous knowledge of active compounds without relying on a ligand complex. Herein we describe pharmacophore virtual screening based on previously reported active molecules that target the toxic RNA that causes myotonic dystrophy type 1 (DM1). DM1-associated defects are caused by sequestration of muscleblind-like 1 protein (MBNL1), an alternative splicing regulator, by expanded CUG repeats (r(CUG)exp). Several small molecules have been found to disrupt the MBNL1-r(CUG)exp complex, ameliorating DM1 defects. Our pharmacophore model identified a number of potential lead compounds from which we selected 11 compounds to evaluate. Of the 11 compounds, several improved DM1 defects both in vitro and in cells.


Assuntos
Distrofia Miotônica/tratamento farmacológico , RNA/genética , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Processamento Alternativo/efeitos dos fármacos , Animais , Desenho Assistido por Computador , Proteínas de Ligação a DNA/metabolismo , Desenho de Fármacos , Humanos , Camundongos , Terapia de Alvo Molecular , Distrofia Miotônica/genética , Distrofia Miotônica/metabolismo , RNA/metabolismo , Proteínas de Ligação a RNA/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA