Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 70(2): 265-273.e8, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29656923

RESUMEN

SF3B is a multi-protein complex essential for branch site (BS) recognition and selection during pre-mRNA splicing. Several splicing modulators with antitumor activity bind SF3B and thereby modulate splicing. Here we report the crystal structure of a human SF3B core in complex with pladienolide B (PB), a macrocyclic splicing modulator and potent inhibitor of tumor cell proliferation. PB stalls SF3B in an open conformation by acting like a wedge within a hinge, modulating SF3B's transition to the closed conformation needed to form the BS adenosine-binding pocket and stably accommodate the BS/U2 duplex. This work explains the structural basis for the splicing modulation activity of PB and related compounds, and reveals key interactions between SF3B and a common pharmacophore, providing a framework for future structure-based drug design.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Epoxi/farmacología , Macrólidos/farmacología , Fosfoproteínas/metabolismo , Factores de Empalme de ARN/metabolismo , Empalme del ARN/efectos de los fármacos , Adenosina/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Sitios de Unión , Proteínas Portadoras/metabolismo , Proliferación Celular/efectos de los fármacos , Diseño de Fármacos , Compuestos Epoxi/química , Compuestos Epoxi/metabolismo , Células HCT116 , Células HeLa , Humanos , Macrólidos/química , Macrólidos/metabolismo , Modelos Moleculares , Complejos Multiproteicos , Fosfoproteínas/química , Fosfoproteínas/genética , Unión Proteica , Conformación Proteica , Precursores del ARN/genética , Precursores del ARN/metabolismo , Factores de Empalme de ARN/química , Factores de Empalme de ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN , Células Sf9 , Relación Estructura-Actividad , Transactivadores
2.
Genes Dev ; 32(3-4): 309-320, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29491137

RESUMEN

Somatic mutations in spliceosome proteins lead to dysregulated RNA splicing and are observed in a variety of cancers. These genetic aberrations may offer a potential intervention point for targeted therapeutics. SF3B1, part of the U2 small nuclear RNP (snRNP), is targeted by splicing modulators, including E7107, the first to enter clinical trials, and, more recently, H3B-8800. Modulating splicing represents a first-in-class opportunity in drug discovery, and elucidating the structural basis for the mode of action opens up new possibilities for structure-based drug design. Here, we present the cryogenic electron microscopy (cryo-EM) structure of the SF3b subcomplex (SF3B1, SF3B3, PHF5A, and SF3B5) bound to E7107 at 3.95 Å. This structure shows that E7107 binds in the branch point adenosine-binding pocket, forming close contacts with key residues that confer resistance upon mutation: SF3B1R1074H and PHF5AY36C The structure suggests a model in which splicing modulators interfere with branch point adenosine recognition and supports a substrate competitive mechanism of action (MOA). Using several related chemical probes, we validate the pose of the compound and support their substrate competitive MOA by comparing their activity against both strong and weak pre-mRNA substrates. Finally, we present functional data and structure-activity relationship (SAR) on the PHF5AR38C mutation that sensitizes cells to some chemical probes but not others. Developing small molecule splicing modulators represents a promising therapeutic approach for a variety of diseases, and this work provides a significant step in enabling structure-based drug design for these elaborate natural products. Importantly, this work also demonstrates that the utilization of cryo-EM in drug discovery is coming of age.


Asunto(s)
Compuestos Epoxi/química , Macrólidos/química , Fosfoproteínas/química , Factores de Empalme de ARN/química , Empalme del ARN/efectos de los fármacos , Empalmosomas/efectos de los fármacos , Proteínas Portadoras/química , Proteínas Portadoras/genética , Proteínas Portadoras/aislamiento & purificación , Microscopía por Crioelectrón , Modelos Moleculares , Mutación , Fosfoproteínas/aislamiento & purificación , Precursores del ARN/metabolismo , Factores de Empalme de ARN/aislamiento & purificación , ARN Mensajero/metabolismo , Proteínas de Unión al ARN , Transactivadores
3.
Biochemistry ; 62(14): 2147-2160, 2023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37403936

RESUMEN

Werner syndrome protein (WRN) is a multifunctional enzyme with helicase, ATPase, and exonuclease activities that are necessary for numerous DNA-related transactions in the human cell. Recent studies identified WRN as a synthetic lethal target in cancers characterized by genomic microsatellite instability resulting from defects in DNA mismatch repair pathways. WRN's helicase activity is essential for the viability of these high microsatellite instability (MSI-H) cancers and thus presents a therapeutic opportunity. To this end, we developed a multiplexed high-throughput screening assay that monitors exonuclease, ATPase, and helicase activities of full-length WRN. This screening campaign led to the discovery of 2-sulfonyl/sulfonamide pyrimidine derivatives as novel covalent inhibitors of WRN helicase activity. The compounds are specific for WRN versus other human RecQ family members and show competitive behavior with ATP. Examination of these novel chemical probes established the sulfonamide NH group as a key driver of compound potency. One of the leading compounds, H3B-960, showed consistent activities in a range of assays (IC50 = 22 nM, KD = 40 nM, KI = 32 nM), and the most potent compound identified, H3B-968, has inhibitory activity IC50 ∼ 10 nM. These kinetic properties trend toward other known covalent druglike molecules. Our work provides a new avenue for screening WRN for inhibitors that may be adaptable to different therapeutic modalities such as targeted protein degradation, as well as a proof of concept for the inhibition of WRN helicase activity by covalent molecules.


Asunto(s)
Neoplasias , Síndrome de Werner , Humanos , Exodesoxirribonucleasas/genética , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , Ensayos Analíticos de Alto Rendimiento , Inestabilidad de Microsatélites , Helicasa del Síndrome de Werner/metabolismo
4.
J Biol Chem ; 298(11): 102539, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36179791

RESUMEN

Recent studies have reported that the peroxisome proliferator-activated receptor gamma (PPARγ) pathway is activated in approximately 40% of patients with muscle-invasive bladder cancer. This led us to investigate pharmacological repression of PPARγ as a possible intervention strategy. Here, we characterize PPARγ antagonists and inverse agonists and find that the former behave as silent ligands, whereas inverse agonists (T0070907 and SR10221) repress downstream PPARγ target genes leading to growth inhibition in bladder cancer cell lines. To understand the mechanism, we determined the ternary crystal structure of PPARγ bound to T0070907 and the corepressor (co-R) peptide NCOR1. The structure shows that the AF-2 helix 12 (H12) rearranges to bind inside the ligand-binding domain, where it forms stabilizing interactions with the compound. This dramatic movement in H12 unveils a large interface for co-R binding. In contrast, the crystal structure of PPARγ bound to a SR10221 analog shows more subtle structural differences, where the compound binds and pushes H12 away from the ligand-binding domain to allow co-R binding. Interestingly, we found that both classes of compound promote recruitment of co-R proteins in biochemical assays but with distinct conformational changes in H12. We validate our structural models using both site-directed mutagenesis and chemical probes. Our findings offer new mechanistic insights into pharmacological modulation of PPARγ signaling.


Asunto(s)
PPAR gamma , Neoplasias de la Vejiga Urinaria , Humanos , PPAR gamma/metabolismo , Ligandos , Benzamidas/farmacología
5.
Subcell Biochem ; 96: 409-432, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33252738

RESUMEN

In this chapter, the essential role of the SF3b multi-protein complex will be discussed in the context of the overall spliceosome. SF3b is critical during spliceosome assembly for recognition of the branch point (BP) adenosine and, by de facto, selection of the 3' splice site. This complex is highly dynamic, undergoing significant conformational changes upon loading of the branch duplex RNA and in its relative positioning during spliceosomal remodeling from the A, pre-B, B, Bact and B* complexes. Ultimately, during the spliceosome activation phase, SF3b must be displaced to unmask the branch point adenosine for the first splicing reaction to occur. In certain cancers, such as the hematological malignancies CML, CLL and MDS, the SF3b subunit SF3B1 is frequently mutated. Recent studies suggest these mutations lead to inappropriate branch point selection and mis-splicing events that appear to be drivers of disease. Finally, the SF3b complex is the target for at least three different classes of natural product-based inhibitors. These inhibitors bind in the BP adenosine-binding pocket and demonstrate a pre-mRNA competitive mechanism of action resulting in either intron retention or exon skipping. These compounds are extremely useful as chemical probes to isolate and characterize early stages of spliceosome assembly. They are also being explored preclinically and clinically as possible agents for hematological cancers.


Asunto(s)
Productos Biológicos/farmacología , Complejos Multiproteicos/antagonistas & inhibidores , Empalmosomas/química , Empalmosomas/efectos de los fármacos , Neoplasias Hematológicas/tratamiento farmacológico , Humanos , Complejos Multiproteicos/genética , Precursores del ARN/genética , Precursores del ARN/metabolismo , Empalme del ARN , Empalmosomas/genética , Empalmosomas/metabolismo
6.
Mol Cell Biochem ; 476(2): 1165-1178, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33188453

RESUMEN

Atrial fibrillation (AF) is the most common cardiac arrhythmia diagnosed in clinical practice. Even though hypertension, congestive heart failure, pulmonary disease, and coronary artery disease are the potential risk factors for AF, the underlying molecular pathology is largely unknown. The reversion of the mature cardiomyocytes to fetal phenotype, impaired ketone body metabolism, mitochondrial dysfunction, and the cellular effect of reactive oxygen species (ROS) are the major underlying biochemical events associated with the molecular pathology of AF. On this background, the present manuscript sheds light into these biochemical events in regard to the metabolic derangements in cardiomyocyte leading to AF, especially with respect to structural, contractile, and electrophysiological properties. In addition, the article critically reviews the current understanding, potential demerits, and translational strategies in the management of AF.


Asunto(s)
Fibrilación Atrial/patología , Feto/fisiopatología , Cuerpos Cetónicos/metabolismo , Mitocondrias/patología , Miocitos Cardíacos/patología , Especies Reactivas de Oxígeno/metabolismo , Fibrilación Atrial/etiología , Fibrilación Atrial/metabolismo , Humanos , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , Fenotipo
7.
Mol Cell Biochem ; 476(9): 3353-3363, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33942219

RESUMEN

Development of incisional hernia (IH) is multifactorial but inflammation and abdominal wall ECM (extracellular matrix) disorganization are key pathological events. We investigated if the differential expression of fibroblast biomarkers reflects the cellular milieu and the dysregulated ECM in IH tissues. Expression of fibroblast biomarkers, including connective tissue growth factor, alpha-smooth muscle actin (α-SMA), CD34 (cluster of differentiation 34), cadherin-11 and fibroblast specific protein 1 (FSP1), was examined by histology and immunofluorescence in the hernial-fascial ring/neck tissue (HRT) and hernia sack tissue (HST) harvested from the patients undergoing hernia surgery and compared with normal fascia (FT) and peritoneum (PT) harvested from brain-dead healthy subjects undergoing organ procurement for transplantation. The H&E staining revealed alterations in tissue architecture, fibroblast morphology, and ECM organization in the IH tissues compared to control. The biomarker for undifferentiated fibroblasts, CD34, was significantly higher in HST and decreased in HRT than the respective FT and PT controls. Also, the findings revealed an increased level of CTGF (connective tissue growth factor) with decrease in α-SMA in both HRT and HST compared to the controls. In addition, an increased level of FSP1 (fibroblast specific protein 1) and cadherin-11 in HRT with decreased level in HST were observed relative to the respective controls (FT and PT). Hence, these findings support the heterogeneity of fibroblast population at the laparotomy site that could contribute to the development of IH. Understanding the mechanisms causing the phenotype switch of these fibroblasts would open novel strategies to prevent the development of IH following laparotomy.


Asunto(s)
Biomarcadores/metabolismo , Matriz Extracelular/patología , Fibroblastos/patología , Hernia Incisional/patología , Fenotipo , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Diferenciación Celular , Matriz Extracelular/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Hernia Incisional/clasificación , Hernia Incisional/metabolismo , Hernia Incisional/cirugía , Masculino , Persona de Mediana Edad , Adulto Joven
8.
Can J Physiol Pharmacol ; 99(1): 36-41, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33049144

RESUMEN

The number of patients diagnosed with atrial fibrillation (AF) has been rising due to increased incidence, enhanced detection methods, and greater survival rates following diagnosis. Due to this increase, AF is now the most commonly diagnosed arrhythmia in clinical practice. AF is characterized by irregular, high-frequency contractions of atrial myocytes that lead to turbulent blood flow and the potential for thrombus formation, stroke, or heart failure. These high-frequency contractions of the atrial myocytes cause an imbalance between metabolic supply and demand. Although advances have been made in understanding the pathophysiology of AF, the etiology and underlying pathogenic mechanism remain unknown. However, recent evidence suggests that cardiomyocyte metabolism involving 5' AMP-activated protein kinase (AMPK) activation is altered in patients with AF. Here, we critically reviewed the current understanding of AMPK activation in AF and how it could affect structural, contractile, and electrophysiological cellular properties in the pathogenesis of AF.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Fibrilación Atrial/patología , Atrios Cardíacos/patología , Miocitos Cardíacos/metabolismo , Potenciales de Acción/fisiología , Animales , Fibrilación Atrial/metabolismo , Fibrilación Atrial/fisiopatología , Modelos Animales de Enfermedad , Atrios Cardíacos/citología , Atrios Cardíacos/metabolismo , Atrios Cardíacos/fisiopatología , Humanos , Contracción Muscular/fisiología
9.
J Biol Chem ; 294(45): 16966-16977, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31582562

RESUMEN

DNMT3A (DNA methyltransferase 3A) is a de novo DNA methyltransferase responsible for establishing CpG methylation patterns within the genome. DNMT3A activity is essential for normal development, and its dysfunction has been linked to developmental disorders and cancer. DNMT3A is frequently mutated in myeloid malignancies with the majority of mutations occurring at Arg-882, where R882H mutations are most frequent. The R882H mutation causes a reduction in DNA methyltransferase activity and hypomethylation at differentially-methylated regions within the genome, ultimately preventing hematopoietic stem cell differentiation and leading to leukemogenesis. Although the means by which the R882H DNMT3A mutation reduces enzymatic activity has been the subject of several studies, the precise mechanism by which this occurs has been elusive. Herein, we demonstrate that in the context of the full-length DNMT3A protein, the R882H mutation stabilizes the formation of large oligomeric DNMT3A species to reduce the overall DNA methyltransferase activity of the mutant protein as well as the WT-R882H complex in a dominant-negative manner. This shift in the DNMT3A oligomeric equilibrium and the resulting reduced enzymatic activity can be partially rescued in the presence of oligomer-disrupting DNMT3L, as well as DNMT3A point mutations along the oligomer-forming interface of the catalytic domain. In addition to modulating the oligomeric state of DNMT3A, the R882H mutation also leads to a DNA-binding defect, which may further reduce enzymatic activity. These findings provide a mechanistic explanation for the observed loss of DNMT3A activity associated with the R882H hot spot mutation in cancer.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/química , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Mutación , Multimerización de Proteína , ADN/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Humanos , Modelos Moleculares , Estructura Cuaternaria de Proteína
10.
Bioorg Med Chem Lett ; 28(8): 1336-1341, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29559278

RESUMEN

The design and synthesis of a novel series of 2,6-disubstituted pyrazine derivatives as CK2 kinase inhibitors is described. Structure-guided optimization of a 5-substituted-3-thiophene carboxylic acid screening hit (3a) led to the development of a lead compound (12b), which shows inhibition in both enzymatic and cellular assays. Subsequent design and hybridization efforts also led to the unexpected identification of analogs with potent PIM kinase activity (14f).


Asunto(s)
Quinasa de la Caseína II/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Pirazinas/farmacología , Línea Celular Tumoral , Diseño de Fármacos , Humanos , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacocinética , Pirazinas/síntesis química , Pirazinas/química , Pirazinas/farmacocinética , Relación Estructura-Actividad
19.
Bioorg Med Chem Lett ; 25(24): 5743-7, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26546219

RESUMEN

The propensity for cancer cells to accumulate additional centrosomes relative to normal cells could be exploited for therapeutic benefit in oncology. Following literature reports that suggested TNKS1 (tankyrase 1) and PARP16 may be involved with spindle structure and function and may play a role in suppressing multi-polar spindle formation in cells with supernumerary centrosomes, we initiated a phenotypic screen to look for small molecule poly (ADP-ribose) polymerase (PARP) enzyme family inhibitors that could produce a multi-polar spindle phenotype via declustering of centrosomes. Screening of AstraZeneca's collection of phthalazinone PARP inhibitors in HeLa cells using high-content screening techniques identified several compounds that produced a multi-polar spindle phenotype at low nanomolar concentrations. Characterization of these compounds across a broad panel of PARP family enzyme assays indicated that they had activity against several PARP family enzymes, including PARP1, 2, 3, 5a, 5b, and 6. Further optimization of these initial hits for improved declustering potency, solubility, permeability, and oral bioavailability resulted in AZ0108, a PARP1, 2, 6 inhibitor that potently inhibits centrosome clustering and is suitable for in vivo efficacy and tolerability studies.


Asunto(s)
Centrosoma/metabolismo , Ftalazinas/química , Inhibidores de Poli(ADP-Ribosa) Polimerasas/química , Administración Oral , Animales , Sitios de Unión , Células CACO-2 , Centrosoma/efectos de los fármacos , Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos , Células HeLa , Humanos , Microsomas/metabolismo , Conformación Molecular , Simulación de Dinámica Molecular , Ftalazinas/administración & dosificación , Ftalazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/administración & dosificación , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Estructura Terciaria de Proteína , Ratas , Tanquirasas/antagonistas & inhibidores , Tanquirasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA