Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 168(1-2): 86-100.e15, 2017 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-27916275

RESUMEN

Type 1 diabetes is characterized by the destruction of pancreatic ß cells, and generating new insulin-producing cells from other cell types is a major aim of regenerative medicine. One promising approach is transdifferentiation of developmentally related pancreatic cell types, including glucagon-producing α cells. In a genetic model, loss of the master regulatory transcription factor Arx is sufficient to induce the conversion of α cells to functional ß-like cells. Here, we identify artemisinins as small molecules that functionally repress Arx by causing its translocation to the cytoplasm. We show that the protein gephyrin is the mammalian target of these antimalarial drugs and that the mechanism of action of these molecules depends on the enhancement of GABAA receptor signaling. Our results in zebrafish, rodents, and primary human pancreatic islets identify gephyrin as a druggable target for the regeneration of pancreatic ß cell mass from α cells.


Asunto(s)
Artemisininas/farmacología , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Modelos Animales de Enfermedad , Receptores de GABA-A/metabolismo , Transducción de Señal , Animales , Arteméter , Artemisininas/administración & dosificación , Proteínas Portadoras/metabolismo , Transdiferenciación Celular/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus Tipo 1/patología , Perfilación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Insulina/genética , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Ratones , Estabilidad Proteica/efectos de los fármacos , Ratas , Análisis de la Célula Individual , Factores de Transcripción/metabolismo , Pez Cebra , Ácido gamma-Aminobutírico/metabolismo
2.
Nat Immunol ; 17(12): 1361-1372, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27798618

RESUMEN

Hemolysis drives susceptibility to bacterial infections and predicts poor outcome from sepsis. These detrimental effects are commonly considered to be a consequence of heme-iron serving as a nutrient for bacteria. We employed a Gram-negative sepsis model and found that elevated heme levels impaired the control of bacterial proliferation independently of heme-iron acquisition by pathogens. Heme strongly inhibited phagocytosis and the migration of human and mouse phagocytes by disrupting actin cytoskeletal dynamics via activation of the GTP-binding Rho family protein Cdc42 by the guanine nucleotide exchange factor DOCK8. A chemical screening approach revealed that quinine effectively prevented heme effects on the cytoskeleton, restored phagocytosis and improved survival in sepsis. These mechanistic insights provide potential therapeutic targets for patients with sepsis or hemolytic disorders.


Asunto(s)
Infecciones por Bacterias Gramnegativas/inmunología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Hemo/metabolismo , Hemólisis/inmunología , Macrófagos/inmunología , Fagocitosis , Sepsis/inmunología , Animales , Antibacterianos/uso terapéutico , Citoesqueleto/metabolismo , Femenino , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Factores de Intercambio de Guanina Nucleótido/genética , Hemo-Oxigenasa 1/genética , Hemólisis/efectos de los fármacos , Humanos , Evasión Inmune , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis/efectos de los fármacos , Quinina/uso terapéutico , Células RAW 264.7 , Sepsis/tratamiento farmacológico , Proteína de Unión al GTP cdc42/metabolismo
3.
Bioorg Med Chem Lett ; 98: 129546, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-37944866

RESUMEN

Epigenetic proteins containing YEATS domains (YD) are an emerging target class in drug discovery. Described herein are the discovery and characterization efforts associated with PFI-6, a new chemical probe for the YD of MLLT1 (ENL/YEATS1) and MLLT3 (AF9/YEATS3). For hit identification, fragment-like mimetics of endogenous YD ligands (crotonylated histone-containing proteins), were synthesized via parallel medicinal chemistry (PMC) and screened for MLLT1 binding. Subsequent SAR studies led to iterative MLLT1/3 binding and selectivity improvements, culminating in the discovery of PFI-6. PFI-6 demonstrates good affinity and selectivity for MLLT1/3 vs. other human YD proteins (YEATS2/4) and engages MLLT3 in cells. Small-molecule X-ray co-crystal structures of two molecules, including PFI-6, bound to the YD of MLLT1/3 are also described. PFI-6 may be a useful tool molecule to better understand the biological effects associated with modulation of MLLT1/3.


Asunto(s)
Histonas , Proteínas Nucleares , Humanos , Proteínas Nucleares/metabolismo , Histonas/metabolismo , Dominios Proteicos , Descubrimiento de Drogas , Proteínas de Neoplasias/metabolismo , Factores de Transcripción/metabolismo
4.
Biol Chem ; 403(4): 391-402, 2022 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-35191283

RESUMEN

Ubiquitination is a key regulatory mechanism vital for maintenance of cellular homeostasis. Protein degradation is induced by E3 ligases via attachment of ubiquitin chains to substrates. Pharmacological exploitation of this phenomenon via targeted protein degradation (TPD) can be achieved with molecular glues or bifunctional molecules facilitating the formation of ternary complexes between an E3 ligase and a given protein of interest (POI), resulting in ubiquitination of the substrate and subsequent proteolysis by the proteasome. Recently, the development of novel covalent fragment screening approaches has enabled the identification of first-in-class ligands for E3 ligases and deubiquitinases revealing so far unexplored binding sites which highlights the potential of these methods to uncover and expand druggable space for new target classes.


Asunto(s)
Complejo de la Endopetidasa Proteasomal , Ubiquitina , Ligandos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
5.
Chembiochem ; 22(12): 2116-2123, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33887086

RESUMEN

Selinexor, a covalent XPO1 inhibitor, is approved in the USA in combination with dexamethasone for penta-refractory multiple myeloma. Additional XPO1 covalent inhibitors are currently in clinical trials for multiple diseases including hematologic malignancies, solid tumor malignancies, glioblastoma multiforme (GBM), and amyotrophic lateral sclerosis (ALS). It is important to measure the target engagement and selectivity of covalent inhibitors to understand the degree of engagement needed for efficacy, while avoiding both mechanism-based and off-target toxicity. Herein, we report clickable probes based on the XPO1 inhibitors selinexor and eltanexor for the labeling of XPO1 in live cells to assess target engagement and selectivity. We used mass spectrometry-based chemoproteomic workflows to profile the proteome-wide selectivity of selinexor and eltanexor and show that they are highly selective for XPO1. Thermal profiling analysis of selinexor further offers an orthogonal approach to measure XPO1 engagement in live cells. We believe these probes and assays will serve as useful tools to further interrogate the biology of XPO1 and its inhibition in cellular and in vivo systems.


Asunto(s)
Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Antineoplásicos/farmacología , Glioblastoma/tratamiento farmacológico , Hidrazinas/farmacología , Carioferinas/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Triazoles/farmacología , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Antineoplásicos/química , Línea Celular Tumoral , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Hidrazinas/química , Carioferinas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Triazoles/química , Proteína Exportina 1
6.
Nat Chem Biol ; 15(8): 822-829, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31285596

RESUMEN

Here, we report the fragment-based discovery of BI-9321, a potent, selective and cellular active antagonist of the NSD3-PWWP1 domain. The human NSD3 protein is encoded by the WHSC1L1 gene located in the 8p11-p12 amplicon, frequently amplified in breast and squamous lung cancer. Recently, it was demonstrated that the PWWP1 domain of NSD3 is required for the viability of acute myeloid leukemia cells. To further elucidate the relevance of NSD3 in cancer biology, we developed a chemical probe, BI-9321, targeting the methyl-lysine binding site of the PWWP1 domain with sub-micromolar in vitro activity and cellular target engagement at 1 µM. As a single agent, BI-9321 downregulates Myc messenger RNA expression and reduces proliferation in MOLM-13 cells. This first-in-class chemical probe BI-9321, together with the negative control BI-9466, will greatly facilitate the elucidation of the underexplored biological function of PWWP domains.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Proteínas Nucleares/antagonistas & inhibidores , Sistemas CRISPR-Cas , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Regulación de la Expresión Génica/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Dominios Proteicos , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo
7.
Nature ; 519(7544): 477-81, 2015 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-25561175

RESUMEN

Cell growth and proliferation are tightly linked to nutrient availability. The mechanistic target of rapamycin complex 1 (mTORC1) integrates the presence of growth factors, energy levels, glucose and amino acids to modulate metabolic status and cellular responses. mTORC1 is activated at the surface of lysosomes by the RAG GTPases and the Ragulator complex through a not fully understood mechanism monitoring amino acid availability in the lysosomal lumen and involving the vacuolar H(+)-ATPase. Here we describe the uncharacterized human member 9 of the solute carrier family 38 (SLC38A9) as a lysosomal membrane-resident protein competent in amino acid transport. Extensive functional proteomic analysis established SLC38A9 as an integral part of the Ragulator-RAG GTPases machinery. Gain of SLC38A9 function rendered cells resistant to amino acid withdrawal, whereas loss of SLC38A9 expression impaired amino-acid-induced mTORC1 activation. Thus SLC38A9 is a physical and functional component of the amino acid sensing machinery that controls the activation of mTOR.


Asunto(s)
Sistemas de Transporte de Aminoácidos/metabolismo , Aminoácidos/metabolismo , Lisosomas/metabolismo , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Línea Celular , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Proteínas de Unión al GTP Monoméricas/metabolismo , Nucleótidos/metabolismo
8.
Nature ; 508(7495): 222-7, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24695225

RESUMEN

Activated RAS GTPase signalling is a critical driver of oncogenic transformation and malignant disease. Cellular models of RAS-dependent cancers have been used to identify experimental small molecules, such as SCH51344, but their molecular mechanism of action remains generally unknown. Here, using a chemical proteomic approach, we identify the target of SCH51344 as the human mutT homologue MTH1 (also known as NUDT1), a nucleotide pool sanitizing enzyme. Loss-of-function of MTH1 impaired growth of KRAS tumour cells, whereas MTH1 overexpression mitigated sensitivity towards SCH51344. Searching for more drug-like inhibitors, we identified the kinase inhibitor crizotinib as a nanomolar suppressor of MTH1 activity. Surprisingly, the clinically used (R)-enantiomer of the drug was inactive, whereas the (S)-enantiomer selectively inhibited MTH1 catalytic activity. Enzymatic assays, chemical proteomic profiling, kinome-wide activity surveys and MTH1 co-crystal structures of both enantiomers provide a rationale for this remarkable stereospecificity. Disruption of nucleotide pool homeostasis via MTH1 inhibition by (S)-crizotinib induced an increase in DNA single-strand breaks, activated DNA repair in human colon carcinoma cells, and effectively suppressed tumour growth in animal models. Our results propose (S)-crizotinib as an attractive chemical entity for further pre-clinical evaluation, and small-molecule inhibitors of MTH1 in general as a promising novel class of anticancer agents.


Asunto(s)
Antineoplásicos/farmacología , Enzimas Reparadoras del ADN/antagonistas & inhibidores , Enzimas Reparadoras del ADN/metabolismo , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Monoéster Fosfórico Hidrolasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Piridinas/farmacología , Aminoquinolinas/farmacología , Animales , Antineoplásicos/química , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Crizotinib , Cristalización , Roturas del ADN de Cadena Simple/efectos de los fármacos , Reparación del ADN , Enzimas Reparadoras del ADN/biosíntesis , Enzimas Reparadoras del ADN/química , Modelos Animales de Enfermedad , Femenino , Homeostasis/efectos de los fármacos , Humanos , Ratones , Ratones SCID , Modelos Moleculares , Nucleótidos/metabolismo , Monoéster Fosfórico Hidrolasas/biosíntesis , Monoéster Fosfórico Hidrolasas/química , Conformación Proteica , Inhibidores de Proteínas Quinasas/química , Proteómica , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , Pirazoles/química , Piridinas/química , Especificidad por Sustrato , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas ras/genética
9.
J Am Chem Soc ; 141(22): 8951-8968, 2019 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-31060360

RESUMEN

Covalent probes can display unmatched potency, selectivity, and duration of action; however, their discovery is challenging. In principle, fragments that can irreversibly bind their target can overcome the low affinity that limits reversible fragment screening, but such electrophilic fragments were considered nonselective and were rarely screened. We hypothesized that mild electrophiles might overcome the selectivity challenge and constructed a library of 993 mildly electrophilic fragments. We characterized this library by a new high-throughput thiol-reactivity assay and screened them against 10 cysteine-containing proteins. Highly reactive and promiscuous fragments were rare and could be easily eliminated. In contrast, we found hits for most targets. Combining our approach with high-throughput crystallography allowed rapid progression to potent and selective probes for two enzymes, the deubiquitinase OTUB2 and the pyrophosphatase NUDT7. No inhibitors were previously known for either. This study highlights the potential of electrophile-fragment screening as a practical and efficient tool for covalent-ligand discovery.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Electrones , Células HEK293 , Humanos , Ligandos , Modelos Moleculares , Peso Molecular , Conformación Proteica , Factores de Tiempo
10.
Nat Chem Biol ; 13(6): 681-690, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28437395

RESUMEN

Small-molecule drugs may complement antibody-based therapies in an immune-oncology setting, yet systematic methods for the identification and characterization of the immunomodulatory properties of these entities are lacking. We surveyed the immumomodulatory potential of 1,402 small chemical molecules, as defined by their ability to alter the cell-cell interactions among peripheral mononuclear leukocytes ex vivo, using automated microscopy and population-wide single-cell image analysis. Unexpectedly, ∼10% of the agents tested affected these cell-cell interactions differentially. The results accurately recapitulated known immunomodulatory drug classes and revealed several clinically approved drugs that unexpectedly harbor the ability to modulate the immune system, which could potentially contribute to their physiological mechanism of action. For instance, the kinase inhibitor crizotinib promoted T cell interactions with monocytes, as well as with cancer cells, through inhibition of the receptor tyrosine kinase MSTR1 and subsequent upregulation of the expression of major histocompatibility complex molecules. The approach offers an attractive platform for the personalized identification and characterization of immunomodulatory therapeutics.


Asunto(s)
Inmunomodulación/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Miosinas Cardíacas/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Crizotinib , Humanos , Cadenas Pesadas de Miosina/metabolismo , Pirazoles/farmacología , Piridinas/farmacología
11.
Angew Chem Int Ed Engl ; 58(2): 515-519, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30431220

RESUMEN

Histone lysine demethylases (KDMs) are involved in the dynamic regulation of gene expression and they play a critical role in several biological processes. Achieving selectivity over the different KDMs has been a major challenge for KDM inhibitor development. Here we report potent and selective KDM5 covalent inhibitors designed to target cysteine residues only present in the KDM5 sub-family. The covalent binding to the targeted proteins was confirmed by MS and time-dependent inhibition. Additional competition assays show that compounds were non 2-OG competitive. Target engagement and ChIP-seq analysis showed that the compounds inhibited the KDM5 members in cells at nano- to micromolar levels and induce a global increase of the H3K4me3 mark at transcriptional start sites.

12.
Nat Methods ; 12(11): 1055-7, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26389571

RESUMEN

Thermal stabilization of proteins after ligand binding provides an efficient means to assess the binding of small molecules to proteins. We show here that in combination with quantitative mass spectrometry, the approach allows for the systematic survey of protein engagement by cellular metabolites and drugs. We profiled the targets of the drugs methotrexate and (S)-crizotinib and the metabolite 2'3'-cGAMP in intact cells and identified the 2'3'-cGAMP cognate transmembrane receptor STING, involved in immune signaling.


Asunto(s)
Proteoma/metabolismo , Pirazoles/química , Piridinas/química , Animales , Proteínas Portadoras/metabolismo , Línea Celular , Línea Celular Tumoral , Biología Computacional , Crizotinib , Diseño de Fármacos , Humanos , Sistema Inmunológico , Células K562 , Ligandos , Espectrometría de Masas , Metotrexato/química , Ratones , Unión Proteica , Inhibidores de Proteínas Quinasas/química , Proteómica , Transducción de Señal , Biología de Sistemas , Temperatura
13.
Nat Chem Biol ; 12(7): 504-10, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27159579

RESUMEN

Bromodomain-containing proteins of the BET family recognize histone lysine acetylation and mediate transcriptional activation of target genes such as the MYC oncogene. Pharmacological inhibitors of BET domains promise therapeutic benefits in a variety of cancers. We performed a high-diversity chemical compound screen for agents capable of modulating BRD4-dependent heterochromatization of a generic reporter in human cells. In addition to known and new compounds targeting BRD4, we identified small molecules that mimic BRD4 inhibition without direct engagement. One such compound was a potent inhibitor of the second bromodomain of TAF1. Using this inhibitor, we discovered that TAF1 synergizes with BRD4 to control proliferation of cancer cells, making TAF1 an attractive epigenetic target in cancers driven by MYC.


Asunto(s)
Cromatina/química , Histona Acetiltransferasas/antagonistas & inhibidores , Histona Acetiltransferasas/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Factores Asociados con la Proteína de Unión a TATA/antagonistas & inhibidores , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Factor de Transcripción TFIID/antagonistas & inhibidores , Factor de Transcripción TFIID/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Proteínas de Ciclo Celular , Línea Celular , Proliferación Celular/efectos de los fármacos , Cromatina/efectos de los fármacos , Cromatina/genética , Cromatina/metabolismo , Histona Acetiltransferasas/química , Humanos , Estructura Molecular , Proteínas Nucleares/química , Dominios Proteicos/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Factores Asociados con la Proteína de Unión a TATA/química , Factor de Transcripción TFIID/química , Factores de Transcripción/química
14.
Angew Chem Int Ed Engl ; 57(50): 16302-16307, 2018 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-30288907

RESUMEN

YEATS domain (YD) containing proteins are an emerging class of epigenetic targets in drug discovery. Dysregulation of these modified lysine-binding proteins has been linked to the onset and progression of cancers. We herein report the discovery and characterisation of the first small-molecule chemical probe, SGC-iMLLT, for the YD of MLLT1 (ENL/YEATS1) and MLLT3 (AF9/YEATS3). SGC-iMLLT is a potent and selective inhibitor of MLLT1/3-histone interactions. Excellent selectivity over other human YD proteins (YEATS2/4) and bromodomains was observed. Furthermore, our probe displays cellular target engagement of MLLT1 and MLLT3. The first small-molecule X-ray co-crystal structures with the MLLT1 YD are also reported. This first-in-class probe molecule can be used to understand MLLT1/3-associated biology and the therapeutic potential of small-molecule YD inhibitors.


Asunto(s)
Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/química , Bibliotecas de Moléculas Pequeñas/química , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/química , Cristalografía por Rayos X , Histonas/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Dominios Proteicos , Mapas de Interacción de Proteínas/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Factores de Transcripción/metabolismo
15.
Nat Chem Biol ; 11(8): 571-578, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26167872

RESUMEN

The CEBPA gene is mutated in 9% of patients with acute myeloid leukemia (AML). Selective expression of a short (30-kDa) CCAAT-enhancer binding protein-α (C/EBPα) translational isoform, termed p30, represents the most common type of CEBPA mutation in AML. The molecular mechanisms underlying p30-mediated transformation remain incompletely understood. We show that C/EBPα p30, but not the normal p42 isoform, preferentially interacts with Wdr5, a key component of SET/MLL (SET-domain/mixed-lineage leukemia) histone-methyltransferase complexes. Accordingly, p30-bound genomic regions were enriched for MLL-dependent H3K4me3 marks. The p30-dependent increase in self-renewal and inhibition of myeloid differentiation required Wdr5, as downregulation of the latter inhibited proliferation and restored differentiation in p30-dependent AML models. OICR-9429 is a new small-molecule antagonist of the Wdr5-MLL interaction. This compound selectively inhibited proliferation and induced differentiation in p30-expressing human AML cells. Our data reveal the mechanism of p30-dependent transformation and establish the essential p30 cofactor Wdr5 as a therapeutic target in CEBPA-mutant AML.


Asunto(s)
Antineoplásicos/farmacología , Compuestos de Bifenilo/farmacología , Dihidropiridinas/farmacología , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Leucemia Mieloide Aguda/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Secuencia de Aminoácidos , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Terapia Molecular Dirigida , Mutación , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Transducción de Señal , Células Tumorales Cultivadas
16.
Bioorg Med Chem ; 25(9): 2657-2665, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28341403

RESUMEN

The PIM family of serine/threonine kinases have become an attractive target for anti-cancer drug development, particularly for certain hematological malignancies. Here, we describe the discovery of a series of inhibitors of the PIM kinase family using a high throughput screening strategy. Through a combination of molecular modeling and optimization studies, the intrinsic potencies and molecular properties of this series of compounds was significantly improved. An excellent pan-PIM isoform inhibition profile was observed across the series, while optimized examples show good selectivity over other kinases. Two PIM-expressing leukemic cancer cell lines, MV4-11 and K562, were employed to evaluate the in vitro anti-proliferative effects of selected inhibitors. Encouraging activities were observed for many examples, with the best example (44) giving an IC50 of 0.75µM against the K562 cell line. These data provide a promising starting point for further development of this series as a new cancer therapy through PIM kinase inhibition.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Rodanina/análogos & derivados , Sulfonamidas/farmacología , Tiazolidinas/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Humanos , Isoenzimas/antagonistas & inhibidores , Células K562 , Ratones , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Simulación del Acoplamiento Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacocinética , Rodanina/síntesis química , Rodanina/farmacocinética , Rodanina/farmacología , Solubilidad , Sulfonamidas/síntesis química , Sulfonamidas/farmacocinética , Tiazolidinas/síntesis química , Tiazolidinas/farmacocinética
17.
Angew Chem Int Ed Engl ; 56(3): 827-831, 2017 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-27966810

RESUMEN

The p300/CBP-associated factor (PCAF) and related GCN5 bromodomain-containing lysine acetyl transferases are members of subfamily I of the bromodomain phylogenetic tree. Iterative cycles of rational inhibitor design and biophysical characterization led to the discovery of the triazolopthalazine-based L-45 (dubbed L-Moses) as the first potent, selective, and cell-active PCAF bromodomain (Brd) inhibitor. Synthesis from readily available (1R,2S)-(-)-norephedrine furnished L-45 in enantiopure form. L-45 was shown to disrupt PCAF-Brd histone H3.3 interaction in cells using a nanoBRET assay, and a co-crystal structure of L-45 with the homologous Brd PfGCN5 from Plasmodium falciparum rationalizes the high selectivity for PCAF and GCN5 bromodomains. Compound L-45 shows no observable cytotoxicity in peripheral blood mononuclear cells (PBMC), good cell-permeability, and metabolic stability in human and mouse liver microsomes, supporting its potential for in vivo use.


Asunto(s)
Compuestos Azo/farmacología , Descubrimiento de Drogas , Hidralazina/farmacología , Sondas Moleculares/farmacología , Factores de Transcripción p300-CBP/antagonistas & inhibidores , Compuestos Azo/síntesis química , Compuestos Azo/química , Relación Dosis-Respuesta a Droga , Hidralazina/síntesis química , Hidralazina/química , Sondas Moleculares/síntesis química , Sondas Moleculares/química , Estructura Molecular , Relación Estructura-Actividad
18.
Nat Chem Biol ; 10(9): 768-773, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25064833

RESUMEN

Genotoxic chemotherapy is the most common cancer treatment strategy. However, its untargeted generic DNA-damaging nature and associated systemic cytotoxicity greatly limit its therapeutic applications. Here, we used a haploid genetic screen in human cells to discover an absolute dependency of the clinically evaluated anticancer compound YM155 on solute carrier family member 35 F2 (SLC35F2), an uncharacterized member of the solute carrier protein family that is highly expressed in a variety of human cancers. YM155 generated DNA damage through intercalation, which was contingent on the expression of SLC35F2 and its drug-importing activity. SLC35F2 expression and YM155 sensitivity correlated across a panel of cancer cell lines, and targeted genome editing verified SLC35F2 as the main determinant of YM155-mediated DNA damage toxicity in vitro and in vivo. These findings suggest a new route to targeted DNA damage by exploiting tumor and patient-specific import of YM155.


Asunto(s)
Daño del ADN/efectos de los fármacos , Imidazoles/farmacología , Sustancias Intercalantes/farmacología , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Naftoquinonas/farmacología , Animales , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , División Celular/fisiología , Línea Celular Tumoral , Supervivencia Celular , Clonación Molecular , Ensayo Cometa , Genoma Humano/efectos de los fármacos , Genoma Humano/genética , Haploidia , Humanos , Imidazoles/metabolismo , Inmunohistoquímica , Ratones , Ratones SCID , Naftoquinonas/metabolismo , ARN Neoplásico/química , ARN Neoplásico/genética
19.
Nat Chem Biol ; 13(2): 133-134, 2017 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-28103226
20.
J Med Chem ; 67(9): 7245-7259, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38635563

RESUMEN

Cofactor mimicry represents an attractive strategy for the development of enzyme inhibitors but can lead to off-target effects due to the evolutionary conservation of binding sites across the proteome. Here, we uncover the ADP-ribose (ADPr) hydrolase NUDT5 as an unexpected, noncovalent, off-target of clinical BTK inhibitors. Using a combination of biochemical, biophysical, and intact cell NanoBRET assays as well as X-ray crystallography, we confirm catalytic inhibition and cellular target engagement of NUDT5 and reveal an unusual binding mode that is independent of the reactive acrylamide warhead. Further investigation of the prototypical BTK inhibitor ibrutinib also revealed potent inhibition of the largely unstudied NUDIX hydrolase family member NUDT14. By exploring structure-activity relationships (SARs) around the core scaffold, we identify a potent, noncovalent, and cell-active dual NUDT5/14 inhibitor. Cocrystallization experiments yielded new insights into the NUDT14 hydrolase active site architecture and inhibitor binding, thus providing a basis for future chemical probe design.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa , Pirofosfatasas , Humanos , Pirofosfatasas/antagonistas & inhibidores , Pirofosfatasas/metabolismo , Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Agammaglobulinemia Tirosina Quinasa/metabolismo , Relación Estructura-Actividad , Cristalografía por Rayos X , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/síntesis química , Pirazoles/química , Pirazoles/farmacología , Pirazoles/síntesis química , Pirazoles/metabolismo , Piperidinas/farmacología , Piperidinas/química , Piperidinas/metabolismo , Piperidinas/síntesis química , Descubrimiento de Drogas , Pirimidinas/química , Pirimidinas/farmacología , Pirimidinas/síntesis química , Pirimidinas/metabolismo , Adenina/análogos & derivados , Adenina/química , Adenina/farmacología , Adenina/metabolismo , Modelos Moleculares , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/síntesis química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA