Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 13(1): 4522, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35927243

RESUMEN

Genomic methods have been valuable for identifying RNA-binding proteins (RBPs) and the genes, pathways, and processes they regulate. Nevertheless, standard motif descriptions cannot be used to predict all RNA targets or test quantitative models for cellular interactions and regulation. We present a complete thermodynamic model for RNA binding to the S. cerevisiae Pumilio protein PUF4 derived from direct binding data for 6180 RNAs measured using the RNA on a massively parallel array (RNA-MaP) platform. The PUF4 model is highly similar to that of the related RBPs, human PUM2 and PUM1, with one marked exception: a single favorable site of base flipping for PUF4, such that PUF4 preferentially binds to a non-contiguous series of residues. These results are foundational for developing and testing cellular models of RNA-RBP interactions and function, for engineering RBPs, for understanding the biophysical nature of RBP binding and the evolutionary landscape of RNAs and RBPs.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Proteínas Fúngicas/metabolismo , Humanos , Unión Proteica , ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Termodinámica
2.
ACS Med Chem Lett ; 12(6): 935-940, 2021 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-34141072

RESUMEN

Myotonic dystrophy type 1 originates from d(CTG·CAG) repeats that undergo aberrant expansion during normal processing because the d(CTG) repeat forms stable hairpin structures. Bidirectional transcription of d(CTG·CAG) yields two RNA transcripts that undergo repeat-associated non-ATG (RAN) translation to form homopolymeric proteins. Thus, both the r(CUG) transcript and the r(CAG) transcript are known to be toxic. We report a pairwise fragment-based, target-guided approach to screen for proximity-induced click dimers formed on the nucleic acid template. This screen uses an azide/alkyne clickable fragment library of nucleic acid-binding ligands incubated in parallel, pairwise reactions as an alternative to our previously reported one-pot screening method. MALDI-TOF mass spectroscopy was used to detect template assisted click products. Hit compounds inhibited the in vitro transcription of d(CTG·CAG)90 bidirectionally with IC50 values in the low micromolar range. This approach may be broadly applicable to other trinucleotide repeat diseases and in targeting other disease-associated nucleic acid sequences.

3.
Biochemistry ; 59(37): 3463-3472, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32856901

RESUMEN

There are few methods available for the rapid discovery of multitarget drugs. Herein, we describe the template-assisted, target-guided discovery of small molecules that recognize d(CTG) in the expanded d(CTG·CAG) sequence and its r(CUG) transcript that cause myotonic dystrophy type 1. A positive cross-selection was performed using a small library of 30 monomeric alkyne- and azide-containing ligands capable of producing >5000 possible di- and trimeric click products. The monomers were incubated with d(CTG)16 or r(CUG)16 under physiological conditions, and both sequences showed selectivity in the proximity-accelerated azide-alkyne [3+2] cycloaddition click reaction. The limited number of click products formed in both selections and the even smaller number of common products suggests that this method is a useful tool for the discovery of single-target and multitarget lead therapeutic agents.


Asunto(s)
ADN/antagonistas & inhibidores , Distrofia Miotónica/tratamiento farmacológico , Distrofia Miotónica/genética , ARN/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Expansión de Repetición de Trinucleótido/efectos de los fármacos , Células Cultivadas , ADN/genética , ADN/metabolismo , Humanos , Distrofia Miotónica/patología , ARN/genética , ARN/metabolismo , Expansión de Repetición de Trinucleótido/genética
4.
J Am Chem Soc ; 142(25): 11165-11172, 2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32478511

RESUMEN

The potent DNA-binding compound triaminotriazine-acridine conjugate (Z1) functions by targeting T:T mismatches in CTG trinucleotide repeats that are responsible for causing neurological diseases such as myotonic dystrophy type 1, but its binding mechanism remains unclear. We solved a crystal structure of Z1 in a complex with DNA containing three consecutive CTG repeats with three T:T mismatches. Crystallographic studies revealed that direct intercalation of two Z1 molecules at both ends of the CTG repeat induces thymine base flipping and DNA backbone deformation to form a four-way junction. The core of the complex unexpectedly adopts a U-shaped head-to-head topology to form a crossover of each chain at the junction site. The crossover junction is held together by two stacked G:C pairs at the central core that rotate with respect to each other in an X-shape to form two nonplanar minor-groove-aligned G·C·G·C tetrads. Two stacked G:C pairs on both sides of the center core are involved in the formation of pseudo-continuous duplex DNA. Four metal-mediated base pairs are observed between the N7 atoms of G and CoII, an interaction that strongly preserves the central junction site. Beyond revealing a new type of ligand-induced, four-way junction, these observations enhance our understanding of the specific supramolecular chemistry of Z1 that is essential for the formation of a noncanonical DNA superstructure. The structural features described here serve as a foundation for the design of new sequence-specific ligands targeting mismatches in the repeat-associated structures.


Asunto(s)
Acridinas/química , ADN/química , Sustancias Intercalantes/química , Triazinas/química , Disparidad de Par Base , Emparejamiento Base , ADN/genética , Conformación de Ácido Nucleico , Timina/química , Repeticiones de Trinucleótidos
5.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 10): 652-656, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31584014

RESUMEN

The structure of a 22-base-pair RNA helix with mismatched pyrimidine base pairs is reported. The helix contains two symmetry-related CUG sequences: a triplet-repeat motif implicated in myotonic dystrophy type 1. The CUG repeat contains a U-U mismatch sandwiched between Watson-Crick pairs. Additionally, the center of the helix contains a dimerized UUCG motif with tandem pyrimidine (U-C/C-U) mismatches flanked by U-G wobble pairs. This region of the structure is significantly different from previously observed structures that share the same sequence and neighboring base pairs. The tandem pyrimidine mismatches are unusual and display sheared, cross-strand stacking geometries that locally constrict the helical width, a type of stacking previously associated with purines in internal loops. Thus, pyrimidine-rich regions of RNA have a high degree of structural diversity.


Asunto(s)
Disparidad de Par Base , Pirimidinas/química , ARN/química , Cristalografía por Rayos X , Enlace de Hidrógeno , Modelos Moleculares , Conformación de Ácido Nucleico , Secuencias Repetitivas de Ácidos Nucleicos
6.
Bioorg Med Chem ; 27(13): 2978-2984, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31113691

RESUMEN

We describe the molecular design, synthesis, and investigation of a series of acridine-triaminotriazine macrocycles that selectively bind to CTG trinucleotide repeats in DNA with minimal nonspecific binding. The limited conformational flexibility enforces the stacking of the triaminotriazine and acridine units. Isothermal titration calorimetry studies and Job plot analyses revealed that the ligands bound to d(CTG) mismatched sites. The acridine and triaminotriazine units were shown to intramolecularly π-stack in aqueous solutions. Compared to a noncyclic analog, the macrocycles showed an almost 10-fold lower cytotoxicity in HeLa cells and up to 4-fold higher transcription inhibition of d(CTG·CAG)74.


Asunto(s)
ADN/química , Repeticiones de Trinucleótidos/genética , Humanos
7.
Proc Natl Acad Sci U S A ; 116(18): 8709-8714, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-30975744

RESUMEN

Developing highly active, multivalent ligands as therapeutic agents is challenging because of delivery issues, limited cell permeability, and toxicity. Here, we report intrinsically cell-penetrating multivalent ligands that target the trinucleotide repeat DNA and RNA in myotonic dystrophy type 1 (DM1), interrupting the disease progression in two ways. The oligomeric ligands are designed based on the repetitive structure of the target with recognition moieties alternating with bisamidinium groove binders to provide an amphiphilic and polycationic structure, mimicking cell-penetrating peptides. Multiple biological studies suggested the success of our multivalency strategy. The designed oligomers maintained cell permeability and exhibited no apparent toxicity both in cells and in mice at working concentrations. Furthermore, the oligomers showed important activities in DM1 cells and in a DM1 liver mouse model, reducing or eliminating prominent DM1 features. Phenotypic recovery of the climbing defect in adult DM1 Drosophila was also observed. This design strategy should be applicable to other repeat expansion diseases and more generally to DNA/RNA-targeted therapeutics.


Asunto(s)
Distrofia Miotónica/tratamiento farmacológico , Proteínas de Unión al ARN/metabolismo , Repeticiones de Trinucleótidos , Animales , ADN , Proteínas de Unión al ADN , Drosophila melanogaster , Células HeLa , Humanos , Ligandos , Hígado/metabolismo , Ratones , Mioblastos/fisiología , Distrofia Miotónica/genética , Proteínas con Motivos de Reconocimiento de ARN , Proteínas de Unión al ARN/química
8.
J Am Chem Soc ; 138(30): 9498-507, 2016 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-27355522

RESUMEN

The construction of a multivalent ligand is an effective way to increase affinity and selectivity toward biomolecular targets with multiple-ligand binding sites. Adopting this strategy, we used a known cell-penetrating peptide (CPP) mimic as a scaffold to develop a series of multivalent ligand constructs that bind to the expanded dCTG (CTG(exp)) and rCUG nucleotide repeats (CUG(exp)) known to cause myotonic dystrophy type I (DM1), an incurable neuromuscular disease. By assembling this polyvalent construct, the hydrophobic ligands are solubilized and delivered into cell nuclei, and their enhanced binding affinity leads to the inhibition of ribonuclear foci formation and a reversal of splicing defects, all at low concentrations. Some of the multivalent ligands are shown to inhibit selectively the in vitro transcription of (CTG·CAG)74, to reduce the concentration of the toxic CUG RNA in DM1 model cells, and to show phenotypic improvement in vivo in a Drosophila model of DM1. This strategy may be useful in drug design for other trinucleotide repeat disorders and more broadly for intracellular multivalent targeting.


Asunto(s)
Péptidos de Penetración Celular/química , Espacio Intracelular/metabolismo , Peptidomiméticos/metabolismo , Animales , Animales Modificados Genéticamente , Transporte Biológico , Drosophila melanogaster/genética , Células HeLa , Humanos , Ligandos , Modelos Moleculares , Conformación de Ácido Nucleico , Peptidomiméticos/química , Conformación Proteica , Repeticiones de Trinucleótidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA