Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Nat Chem Biol ; 9(3): 184-91, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23292653

RESUMEN

We describe the discovery of UNC1215, a potent and selective chemical probe for the methyllysine (Kme) reading function of L3MBTL3, a member of the malignant brain tumor (MBT) family of chromatin-interacting transcriptional repressors. UNC1215 binds L3MBTL3 with a K(d) of 120 nM, competitively displacing mono- or dimethyllysine-containing peptides, and is greater than 50-fold more potent toward L3MBTL3 than other members of the MBT family while also demonstrating selectivity against more than 200 other reader domains examined. X-ray crystallography identified a unique 2:2 polyvalent mode of interaction between UNC1215 and L3MBTL3. In cells, UNC1215 is nontoxic and directly binds L3MBTL3 via the Kme-binding pocket of the MBT domains. UNC1215 increases the cellular mobility of GFP-L3MBTL3 fusion proteins, and point mutants that disrupt the Kme-binding function of GFP-L3MBTL3 phenocopy the effects of UNC1215 on localization. Finally, UNC1215 was used to reveal a new Kme-dependent interaction of L3MBTL3 with BCLAF1, a protein implicated in DNA damage repair and apoptosis.


Asunto(s)
Benzamidas/farmacología , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Descubrimiento de Drogas , Lisina/análogos & derivados , Sondas Moleculares/farmacología , Piperidinas/farmacología , Benzamidas/química , Benzamidas/metabolismo , Unión Competitiva/efectos de los fármacos , Cristalografía por Rayos X , Proteínas de Unión al ADN/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Lisina/antagonistas & inhibidores , Lisina/química , Lisina/metabolismo , Modelos Moleculares , Sondas Moleculares/química , Sondas Moleculares/metabolismo , Estructura Molecular , Piperidinas/química , Piperidinas/metabolismo , Estructura Terciaria de Proteína , Proteínas Represoras/metabolismo , Relación Estructura-Actividad , Proteínas Supresoras de Tumor/metabolismo
2.
J Med Chem ; 66(20): 14335-14356, 2023 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-37823891

RESUMEN

Tyrosine kinase 2 (TYK2) is a nonreceptor tyrosine kinase that belongs to the JAK family also comprising JAK1, JAK2, and JAK3. TYK2 is an attractive target for various autoimmune diseases as it regulates signal transduction downstream of IL-23 and IL-12 receptors. Selective TYK2 inhibition offers a differentiated clinical profile compared to currently approved JAK inhibitors. However, selectivity for TYK2 versus other JAK family members has been difficult to achieve with small molecules that inhibit the catalytically active kinase domain. Successful targeting of the TYK2 pseudokinase domain as a strategy to achieve isoform selectivity was recently exemplified with deucravacitinib. Described herein is the optimization of selective TYK2 inhibitors targeting the pseudokinase domain, resulting in the discovery of the clinical candidate ABBV-712 (21).


Asunto(s)
Enfermedades Autoinmunes , TYK2 Quinasa , Humanos , Quinasas Janus
3.
J Comput Chem ; 33(6): 659-65, 2012 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-22183769

RESUMEN

Methyllysine histone code readers constitute a new promising group of potential drug targets. For instance, L3MBTL1, a malignant brain tumor (MBT) protein, selectively binds mono- and di-methyllysine epigenetic marks (KMe, KMe(2) ) that eventually results in the negative regulation of multiple genes through the E2F/Rb oncogenic pathway. There is a pressing need in potent and selective small-molecule probes that would enable further target validation and might become therapeutic leads. Such an endeavor would require efficient tools to assess the free energy of protein-ligand binding. However, due to an unparalleled function of the MBT binding pocket (i.e., selective binding to KMe/KMe(2) ) and because of its distinctive structure representing a small aromatic "cage," an accurate assessment of its binding affinity to a ligand appears to be a challenging task. Here, we report a comparative analysis of computationally affordable affinity predictors applied to a set of seven small-molecule ligands interacting with L3MBTL1. The analysis deals with novel ligands and targets, but applies widespread computational approaches and intuitive comparison metrics that makes this study compatible with and incremental to earlier large scale accounts on the efficiency of affinity predictors. Ultimately, this study has revealed three top performers, far ahead of the other techniques, including two scoring functions, PMF04 and PLP, along with a simulation-based method MM-PB/SA. We discuss why some methods may perform better than others on this target class, the limits of their application, as well as how the efficiency of the most CPU-demanding techniques could be optimized.


Asunto(s)
Código de Histonas , Ligandos , Lisina/análogos & derivados , Simulación de Dinámica Molecular , Lisina/química , Modelos Moleculares , Termodinámica
4.
J Am Chem Soc ; 133(14): 5357-62, 2011 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-21428286

RESUMEN

Histone lysine methylation (Kme) encodes essential information modulating many biological processes including gene expression and transcriptional regulation. However, the atomic-level recognition mechanisms of methylated histones by their respective adaptor proteins are still elusive. For instance, it is unclear how L3MBTL1, a methyl-lysine histone code reader, recognizes equally well both mono- and dimethyl marks but ignores unmodified and trimethylated lysine residues. We made use of molecular dynamics (MD) and free energy perturbation (FEP) techniques in order to investigate the energetics and dynamics of the methyl-lysine recognition. Isothermal titration calorimetry (ITC) was employed to experimentally validate the computational findings. Both computational and experimental methods were applied to a set of designed "biophysical" probes that mimic the shape of a single lysine residue and reproduce the binding affinities of cognate histone peptides. Our results suggest that, besides forming favorable interactions, the L3MBTL1 binding pocket energetically penalizes both methylation states and has most probably evolved as a "compromise" that nonoptimally fits to both mono- and dimethyl-lysine marks.


Asunto(s)
Lisina/química , Lisina/metabolismo , Simulación de Dinámica Molecular , Sondas Moleculares/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Sitios de Unión , Materiales Biomiméticos/química , Materiales Biomiméticos/metabolismo , Histonas/química , Histonas/metabolismo , Metilación , Sondas Moleculares/química , Conformación Proteica , Estructura Terciaria de Proteína , Termodinámica
5.
Sci Rep ; 9(1): 9089, 2019 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-31235749

RESUMEN

IL-36 cytokines are pro-inflammatory members of the IL-1 family that are upregulated in inflammatory disorders. Specifically, IL-36γ is highly expressed in active psoriatic lesions and can drive pro-inflammatory processes in 3D human skin equivalents supporting a role for this target in skin inflammation. Small molecule antagonists of interleukins have been historically challenging to generate. Nevertheless, we performed a small molecule high-throughput screen to identify IL-36 antagonists using a novel TR-FRET binding assay. Several compounds, including 2-oxypyrimidine containing structural analogs of the marketed endothelin receptor A antagonist Ambrisentan, were identified as hits from the screen. A-552 was identified as a the most potent antagonist of human IL-36γ, but not the closely related family member IL-36α, was capable of attenuating IL-36γ induced responses in mouse and human disease models. Additionally, x-ray crystallography studies identified key amino acid residues in the binding pocket present in human IL-36γ that are absent in human IL-36α. A-552 represents a first-in-class small molecule antagonist of IL-36 signaling that could be used as a chemical tool to further investigate the role of this pathway in inflammatory skin diseases such as psoriasis.


Asunto(s)
Interleucina-1/antagonistas & inhibidores , Psoriasis/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Psoriasis/metabolismo , Psoriasis/patología , Piel/efectos de los fármacos , Piel/patología , Bibliotecas de Moléculas Pequeñas/uso terapéutico
6.
J Med Chem ; 57(15): 6822-33, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-25032507

RESUMEN

The lysine methyltransferase SETD8 is the only known methyltransferase that catalyzes monomethylation of histone H4 lysine 20 (H4K20). Monomethylation of H4K20 has been implicated in regulating diverse biological processes including the DNA damage response. In addition to H4K20, SETD8 monomethylates non-histone substrates including proliferating cell nuclear antigen (PCNA) and promotes carcinogenesis by deregulating PCNA expression. However, selective inhibitors of SETD8 are scarce. The only known selective inhibitor of SETD8 to date is nahuoic acid A, a marine natural product, which is competitive with the cofactor. Here, we report the discovery of the first substrate-competitive inhibitor of SETD8, UNC0379 (1). This small-molecule inhibitor is active in multiple biochemical assays. Its affinity to SETD8 was confirmed by ITC (isothermal titration calorimetry) and SPR (surface plasmon resonance) studies. Importantly, compound 1 is selective for SETD8 over 15 other methyltransferases. We also describe structure-activity relationships (SAR) of this series.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Pirrolidinas/química , Quinazolinas/química , Calorimetría , N-Metiltransferasa de Histona-Lisina/química , Pirrolidinas/síntesis química , Quinazolinas/síntesis química , Relación Estructura-Actividad , Resonancia por Plasmón de Superficie
7.
J Med Chem ; 56(18): 7358-71, 2013 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-24040942

RESUMEN

Lysine methylation is a key epigenetic mark, the dysregulation of which is linked to many diseases. Small-molecule antagonism of methyl-lysine (Kme) binding proteins that recognize such epigenetic marks can improve our understanding of these regulatory mechanisms and potentially validate Kme binding proteins as drug-discovery targets. We previously reported the discovery of 1 (UNC1215), the first potent and selective small-molecule chemical probe of a methyl-lysine reader protein, L3MBTL3, which antagonizes the mono- and dimethyl-lysine reading function of L3MBTL3. The design, synthesis, and structure-activity relationship studies that led to the discovery of 1 are described herein. These efforts established the requirements for potent L3MBTL3 binding and enabled the design of novel antagonists, such as compound 2 (UNC1679), that maintain in vitro and cellular potency with improved selectivity against other MBT-containing proteins. The antagonists described were also found to effectively interact with unlabeled endogenous L3MBTL3 in cells.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Lisina/metabolismo , Bibliotecas de Moléculas Pequeñas/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/química , Diseño de Fármacos , Células HEK293 , Humanos , Concentración 50 Inhibidora , Ligandos , Modelos Moleculares , Estructura Terciaria de Proteína , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad , Especificidad por Sustrato
8.
Curr Chem Genomics ; 5: 51-61, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22145013

RESUMEN

The recognition of methyl-lysine and -arginine residues on both histone and other proteins by specific "reader" elements is important for chromatin regulation, gene expression, and control of cell-cycle progression. Recently the crucial role of these reader proteins in cancer development and dedifferentiation has emerged, owing to the increased interest among the scientific community. The methyl-lysine and -arginine readers are a large and very diverse set of effector proteins and targeting them with small molecule probes in drug discovery will inevitably require a detailed understanding of their structural biology and mechanism of binding. In the following review, the critical elements of methyl-lysine and -arginine recognition will be summarized with respect to each protein family and initial results in assay development, probe design, and drug discovery will be highlighted.

9.
J Med Chem ; 54(17): 6139-50, 2011 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-21780790

RESUMEN

Protein lysine methyltransferase G9a plays key roles in the transcriptional repression of a variety of genes via dimethylation of lysine 9 on histone H3 (H3K9me2) of chromatin as well as dimethylation of nonhistone proteins including tumor suppressor p53. We previously reported the discovery of UNC0321 (3), the most potent G9a inhibitor to date, via structure-based design and structure-activity relationship (SAR) exploration of the quinazoline scaffold represented by BIX01294 (1). Despite its very high in vitro potency, compound 3 lacks sufficient cellular potency. The design and synthesis of several generations of new analogues aimed at improving cell membrane permeability while maintaining high in vitro potency resulted in the discovery of a number of novel G9a inhibitors such as UNC0646 (6) and UNC0631 (7) with excellent potency in a variety of cell lines and excellent separation of functional potency versus cell toxicity. The design, synthesis, and cellular SAR of these potent G9a inhibitors are described.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Neoplasias de la Próstata/tratamiento farmacológico , Quinazolinas/farmacología , Western Blotting , Neoplasias de la Mama/enzimología , Neoplasias del Colon/enzimología , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Femenino , Antígenos de Histocompatibilidad , Humanos , Masculino , Modelos Moleculares , Neoplasias de la Próstata/enzimología , Unión Proteica , Conformación Proteica , Quinazolinas/síntesis química , Quinazolinas/química , Relación Estructura-Actividad , Células Tumorales Cultivadas
10.
J Med Chem ; 54(7): 2504-11, 2011 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-21417280

RESUMEN

Proteins which bind methylated lysines ("readers" of the histone code) are important components in the epigenetic regulation of gene expression and can also modulate other proteins that contain methyl-lysine such as p53 and Rb. Recognition of methyl-lysine marks by MBT domains leads to compaction of chromatin and a repressed transcriptional state. Antagonists of MBT domains would serve as probes to interrogate the functional role of these proteins and initiate the chemical biology of methyl-lysine readers as a target class. Small-molecule MBT antagonists were designed based on the structure of histone peptide-MBT complexes and their interaction with MBT domains determined using a chemiluminescent assay and ITC. The ligands discovered antagonize native histone peptide binding, exhibiting 5-fold stronger binding affinity to L3MBTL1 than its preferred histone peptide. The first cocrystal structure of a small molecule bound to L3MBTL1 was determined and provides new insights into binding requirements for further ligand design.


Asunto(s)
Lisina/metabolismo , Proteínas Nucleares/metabolismo , Bibliotecas de Moléculas Pequeñas/metabolismo , Sitios de Unión , Calorimetría , Descubrimiento de Drogas , Ensayos Analíticos de Alto Rendimiento , Humanos , Ligandos , Mediciones Luminiscentes , Metilación , Modelos Moleculares , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/química , Peptidomiméticos/química , Peptidomiméticos/metabolismo , Peptidomiméticos/farmacología , Estructura Terciaria de Proteína , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología
11.
J Med Chem ; 53(21): 7625-31, 2010 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-20931980

RESUMEN

The malignant brain tumor (MBT) repeat is an important epigenetic-code "reader" and is functionally associated with differentiation, gene silencing, and tumor suppression. (1-3) Small molecule probes of MBT domains should enable a systematic study of MBT-containing proteins and potentially reveal novel druggable targets. We designed and applied a virtual screening strategy that identified potential MBT antagonists in a large database of commercially available compounds. A small set of virtual hits was purchased and submitted to experimental testing. Nineteen of the purchased compounds showed a specific dose-dependent protein binding and will provide critical structure-activity information for subsequent lead generation and optimization.


Asunto(s)
Antineoplásicos/química , Proteínas de Neoplasias/química , Relación Estructura-Actividad Cuantitativa , Proteínas Cromosómicas no Histona , Bases de Datos Factuales , Epigénesis Genética , Ensayos Analíticos de Alto Rendimiento , Humanos , Modelos Moleculares , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Estructura Terciaria de Proteína , Proteínas Represoras , Estereoisomerismo , Proteínas Supresoras de Tumor
12.
J Biomol Screen ; 15(1): 62-71, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20008125

RESUMEN

The histone code comprises many posttranslational modifications that occur mainly in histone tail peptides. The identity and location of these marks are read by a variety of histone-binding proteins that are emerging as important regulators of cellular differentiation and development and are increasingly being implicated in numerous disease states. The authors describe the development of the first high-throughput screening assay for the discovery of inhibitors of methyl-lysine binding proteins that will be used to initiate a full-scale discovery effort for this broad target class. They focus on the development of an AlphaScreen-based assay for malignant brain tumor (MBT) domain-containing proteins, which bind to the lower methylation states of lysine residues present in histone tail peptides. This assay takes advantage of the avidity of the AlphaScreen beads to clear the hurdle to assay development presented by the low micromolar binding constants of the histone binding proteins for their cognate peptides. The assay is applicable to other families of methyl-lysine binding proteins, and it has the potential to be used in screening efforts toward the discovery of novel small molecules with utility as research tools for cellular reprogramming and ultimately drug discovery.


Asunto(s)
Bioensayo/métodos , Epigénesis Genética , Lisina/metabolismo , Péptidos/metabolismo , Proteínas/metabolismo , Bibliotecas de Moléculas Pequeñas/análisis , Bibliotecas de Moléculas Pequeñas/farmacología , Automatización , Unión Competitiva/efectos de los fármacos , Tampones (Química) , Epigénesis Genética/efectos de los fármacos , Histonas/metabolismo , Ligandos , Metilación/efectos de los fármacos , Miniaturización , Proteínas de Neoplasias/metabolismo , Proyectos Piloto , Polisorbatos , Unión Proteica/efectos de los fármacos , Cloruro de Sodio , Factores de Tiempo , Volumetría
13.
J Med Chem ; 53(15): 5844-57, 2010 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-20614940

RESUMEN

Protein lysine methyltransferase G9a, which catalyzes methylation of lysine 9 of histone H3 (H3K9) and lysine 373 (K373) of p53, is overexpressed in human cancers. Genetic knockdown of G9a inhibits cancer cell growth, and the dimethylation of p53 K373 results in the inactivation of p53. Initial SAR exploration of the 2,4-diamino-6,7-dimethoxyquinazoline template represented by 3a (BIX01294), a selective small molecule inhibitor of G9a and GLP, led to the discovery of 10 (UNC0224) as a potent G9a inhibitor with excellent selectivity. A high resolution X-ray crystal structure of the G9a-10 complex, the first cocrystal structure of G9a with a small molecule inhibitor, was obtained. On the basis of the structural insights revealed by this cocrystal structure, optimization of the 7-dimethylaminopropoxy side chain of 10 resulted in the discovery of 29 (UNC0321) (Morrison K(i) = 63 pM), which is the first G9a inhibitor with picomolar potency and the most potent G9a inhibitor to date.


Asunto(s)
Azepinas/síntesis química , Antígenos de Histocompatibilidad/química , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/química , Modelos Moleculares , Quinazolinas/síntesis química , Azepinas/química , Cristalografía por Rayos X , Diseño de Fármacos , Humanos , Unión Proteica , Conformación Proteica , Quinazolinas/química , Relación Estructura-Actividad
14.
J Med Chem ; 52(24): 7950-3, 2009 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-19891491

RESUMEN

SAR exploration of the 2,4-diamino-6,7-dimethoxyquinazoline template led to the discovery of 8 (UNC0224) as a potent and selective G9a inhibitor. A high resolution X-ray crystal structure of the G9a-8 complex, the first cocrystal structure of G9a with a small molecule inhibitor, was obtained. The cocrystal structure validated our binding hypothesis and will enable structure-based design of novel inhibitors. 8 is a useful tool for investigating the biology of G9a and its roles in chromatin remodeling.


Asunto(s)
Inhibidores Enzimáticos/farmacología , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Quinazolinas/farmacología , Cristalografía por Rayos X , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/metabolismo , Modelos Moleculares , Quinazolinas/síntesis química , Quinazolinas/química , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA