Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros











Intervalo de año de publicación
1.
Commun Chem ; 7(1): 168, 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39085342

RESUMEN

Fragment screening is a popular strategy of generating viable chemical starting points especially for challenging targets. Although fragments provide a better coverage of chemical space and they have typically higher chance of binding, their weak affinity necessitates highly sensitive biophysical assays. Here, we introduce a screening concept that combines evolutionary optimized fragment pharmacophores with the use of a photoaffinity handle that enables high hit rates by LC-MS-based detection. The sensitivity of our screening protocol was further improved by a target-conjugated photocatalyst. We have designed, synthesized, and screened 100 diazirine-tagged fragments against three benchmark and three therapeutically relevant protein targets of different tractability. Our therapeutic targets included a conventional enzyme, the first bromodomain of BRD4, a protein-protein interaction represented by the oncogenic KRasG12D protein, and the yet unliganded N-terminal domain of the STAT5B transcription factor. We have discovered several fragment hits against all three targets and identified their binding sites via enzymatic digestion, structural studies and modeling. Our results revealed that this protocol outperforms screening traditional fully functionalized and photoaffinity fragments in better exploration of the available binding sites and higher hit rates observed for even difficult targets.

2.
Cancer Discov ; 14(8): 1457-1475, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-38587317

RESUMEN

Microsatellite-unstable (MSI) cancers require WRN helicase to resolve replication stress due to expanded DNA (TA)n dinucleotide repeats. WRN is a promising synthetic lethal target for MSI tumors, and WRN inhibitors are in development. In this study, we used CRISPR-Cas9 base editing to map WRN residues critical for MSI cells, validating the helicase domain as the primary drug target. Fragment-based screening led to the development of potent and highly selective WRN helicase covalent inhibitors. These compounds selectively suppressed MSI model growth in vitro and in vivo by mimicking WRN loss, inducing DNA double-strand breaks at expanded TA repeats and DNA damage. Assessment of biomarkers in preclinical models linked TA-repeat expansions and mismatch repair alterations to compound activity. Efficacy was confirmed in immunotherapy-resistant organoids and patient-derived xenograft models. The discovery of potent, selective covalent WRN inhibitors provides proof of concept for synthetic lethal targeting of WRN in MSI cancer and tools to dissect WRN biology. Significance: We report the discovery and characterization of potent, selective WRN helicase inhibitors for MSI cancer treatment, with biomarker analysis and evaluation of efficacy in vivo and in immunotherapy-refractory preclinical models. These findings pave the way to translate WRN inhibition into MSI cancer therapies and provide tools to investigate WRN biology. See related commentary by Wainberg, p. 1369.


Asunto(s)
Helicasa del Síndrome de Werner , Humanos , Helicasa del Síndrome de Werner/genética , Ratones , Animales , Inestabilidad de Microsatélites , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Línea Celular Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico
3.
ACS Infect Dis ; 9(11): 2340-2357, 2023 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-37906637

RESUMEN

Leishmaniases are a collection of neglected tropical diseases caused by kinetoplastid parasites in the genus Leishmania. Current chemotherapies are severely limited, and the need for new antileishmanials is of pressing international importance. Bromodomains are epigenetic reader domains that have shown promising therapeutic potential for cancer therapy and may also present an attractive target to treat parasitic diseases. Here, we investigate Leishmania donovani bromodomain factor 5 (LdBDF5) as a target for antileishmanial drug discovery. LdBDF5 contains a pair of bromodomains (BD5.1 and BD5.2) in an N-terminal tandem repeat. We purified recombinant bromodomains of L. donovani BDF5 and determined the structure of BD5.2 by X-ray crystallography. Using a histone peptide microarray and fluorescence polarization assay, we identified binding interactions of LdBDF5 bromodomains with acetylated peptides derived from histones H2B and H4. In orthogonal biophysical assays including thermal shift assays, fluorescence polarization, and NMR, we showed that BDF5 bromodomains bind to human bromodomain inhibitors SGC-CBP30, bromosporine, and I-BRD9; moreover, SGC-CBP30 exhibited activity against Leishmania promastigotes in cell viability assays. These findings exemplify the potential BDF5 holds as a possible drug target in Leishmania and provide a foundation for the future development of optimized antileishmanial compounds targeting this epigenetic reader protein.


Asunto(s)
Antiprotozoarios , Factor V , Humanos , Factor V/metabolismo , Histonas/química , Histonas/metabolismo , Dominios Proteicos , Antiprotozoarios/farmacología , Descubrimiento de Drogas , Factores de Transcripción/metabolismo
4.
RSC Med Chem ; 14(4): 671-679, 2023 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-37122547

RESUMEN

The screening of covalent or 'reactive' fragment libraries against proteins is becoming an integral approach in hit identification, enabling the development of targeted covalent inhibitors and tools. To date, reactive fragment screening has been limited to targeting cysteine residues, thus restricting applicability across the proteome. Carboxylate residues present a unique opportunity to expand the accessible residues due to high proteome occurrence (∼12%). Herein, we present the development of a carboxylate-targeting reactive fragment screening platform utilising 2-aryl-5-carboxytetrazole (ACT) as the photoreactive functionality. The utility of ACT photoreactive fragments (ACT-PhABits) was evaluated by screening a 546-membered library with a small panel of purified proteins. Hits identified for BCL6 and KRASG12D were characterised by LC-MS/MS studies, revealing the selectivity of the ACT group. Finally, a photosensitised approach to ACT activation was developed, obviating the need for high energy UV-B light.

5.
ACS Chem Biol ; 18(9): 1926-1937, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37084287

RESUMEN

Sulfur(VI) fluorides (SFs) have emerged as valuable electrophiles for the design of "beyond-cysteine" covalent inhibitors and offer potential for expansion of the liganded proteome. Since SFs target a broad range of nucleophilic amino acids, they deliver an approach for the covalent modification of proteins without requirement for a proximal cysteine residue. Further to this, libraries of reactive fragments present an innovative approach for the discovery of ligands and tools for proteins of interest by leveraging a breadth of mass spectrometry analytical approaches. Herein, we report a screening approach that exploits the unique properties of SFs for this purpose. Libraries of SF-containing reactive fragments were synthesized, and a direct-to-biology workflow was taken to efficiently identify hit compounds for CAII and BCL6. The most promising hits were further characterized to establish the site(s) of covalent modification, modification kinetics, and target engagement in cells. Crystallography was used to gain a detailed molecular understanding of how these reactive fragments bind to their target. It is anticipated that this screening protocol can be used for the accelerated discovery of "beyond-cysteine" covalent inhibitors.


Asunto(s)
Cisteína , Fluoruros , Cisteína/química , Ligandos , Aminoácidos , Azufre
6.
J Org Chem ; 87(7): 4603-4616, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35302774

RESUMEN

A modular approach to prepare tri- and tetracyclic carbazoles by a sequential [3 + 2]heteroannulation is described. First, optimization of Pd-catalyzed Buchwald-Hartwig amination followed by C/N-arylation in a one-pot process is established. Second, mechanistic analyses identified the origins of chemo- and regioselective sequential control of both bond-forming steps. Finally, the substrate scope is demonstrated by the preparation of a range of tri- and tetracyclic carbazoles, including expedient access to several natural products and anti-cancer agents.


Asunto(s)
Carbazoles , Paladio , Aminación , Catálisis
7.
ACS Chem Biol ; 16(10): 1961-1967, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33835779

RESUMEN

Metabolic oligosaccharide engineering (MOE) has fundamentally contributed to our understanding of protein glycosylation. Efficient MOE reagents are activated into nucleotide-sugars by cellular biosynthetic machineries, introduced into glycoproteins and traceable by bioorthogonal chemistry. Despite their widespread use, the metabolic fate of many MOE reagents is only beginning to be mapped. While metabolic interconnectivity can affect probe specificity, poor uptake by biosynthetic salvage pathways may impact probe sensitivity and trigger side reactions. Here, we use metabolic engineering to turn the weak alkyne-tagged MOE reagents Ac4GalNAlk and Ac4GlcNAlk into efficient chemical tools to probe protein glycosylation. We find that bypassing a metabolic bottleneck with an engineered version of the pyrophosphorylase AGX1 boosts nucleotide-sugar biosynthesis and increases bioorthogonal cell surface labeling by up to two orders of magnitude. A comparison with known azide-tagged MOE reagents reveals major differences in glycoprotein labeling, substantially expanding the toolbox of chemical glycobiology.


Asunto(s)
Galactosamina/análogos & derivados , Galactosamina/metabolismo , Galactosiltransferasas/metabolismo , Glucosamina/análogos & derivados , Glucosamina/metabolismo , Alquinos/química , Secuencia de Aminoácidos , Animales , Azidas/química , Línea Celular Tumoral , Química Clic , Colorantes Fluorescentes/química , Glicoproteínas/química , Glicoproteínas/metabolismo , Glicosilación , Humanos , Ingeniería Metabólica/métodos , Ratones , Sondas Moleculares/química , Oligosacáridos/biosíntesis , Polisacáridos/biosíntesis , Azúcares de Uridina Difosfato/biosíntesis , Azúcares de Uridina Difosfato/metabolismo
8.
Angew Chem Int Ed Engl ; 59(47): 21096-21105, 2020 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-32745361

RESUMEN

Advances in genomic analyses enable the identification of new proteins that are associated with disease. To validate these targets, tool molecules are required to demonstrate that a ligand can have a disease-modifying effect. Currently, as tools are reported for only a fraction of the proteome, platforms for ligand discovery are essential to leverage insights from genomic analyses. Fragment screening offers an efficient approach to explore chemical space. Presented here is a fragment-screening platform, termed PhABits (PhotoAffinity Bits), which utilizes a library of photoreactive fragments to covalently capture fragment-protein interactions. Hits can be profiled to determine potency and the site of crosslinking, and subsequently developed as reporters in a competitive displacement assay to identify novel hit matter. The PhABit platform is envisioned to be widely applicable to novel protein targets, identifying starting points in the development of therapeutics.


Asunto(s)
Antineoplásicos/análisis , Compuestos Bicíclicos Heterocíclicos con Puentes/análisis , Reactivos de Enlaces Cruzados/química , Etiquetas de Fotoafinidad/química , Pirazoles/análisis , Quinoxalinas/análisis , Sulfonamidas/análisis , Vemurafenib/análisis , Antineoplásicos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Humanos , Ligandos , Estructura Molecular , Proteínas/antagonistas & inhibidores , Proteínas/química , Pirazoles/farmacología , Quinoxalinas/farmacología , Sulfonamidas/farmacología , Vemurafenib/farmacología
9.
Chembiochem ; 21(11): 1647-1655, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-31919953

RESUMEN

The hypoxia-inducible factors (HIFs) are key transcription factors in determining cellular responses involving alterations in protein levels in response to limited oxygen availability in animal cells. 2-Oxoglutarate-dependent oxygenases play key roles in regulating levels of HIF and its transcriptional activity. We describe MS-based proteomics studies in which we compared the results of subjecting human breast cancer MCF-7 cells to hypoxia or treating them with a cell-penetrating derivative (dimethyl N-oxalylglycine; DMOG) of the stable 2OG analogue N-oxalylglycine. The proteomic results are consistent with reported transcriptomic analyses and support the proposed key roles of 2OG-dependent HIF prolyl- and asparaginyl-hydroxylases in the hypoxic response. Differences between the data sets for hypoxia and DMOG might reflect context-dependent effects or HIF-independent effects of DMOG.


Asunto(s)
Aminoácidos Dicarboxílicos/farmacología , Hipoxia de la Célula/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Oxígeno/farmacología , Proteoma/genética , Transcriptoma , Atlas como Asunto , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ontología de Genes , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Células MCF-7 , Redes y Vías Metabólicas/genética , Anotación de Secuencia Molecular , Proteoma/clasificación , Proteoma/metabolismo , Proteómica/métodos
10.
Angew Chem Int Ed Engl ; 58(48): 17322-17327, 2019 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-31518032

RESUMEN

The CDK family plays a crucial role in the control of the cell cycle. Dysregulation and mutation of the CDKs has been implicated in cancer and the CDKs have been investigated extensively as potential therapeutic targets. Selective inhibition of specific isoforms of the CDKs is crucial to achieve therapeutic effect while minimising toxicity. We present a group of photoaffinity probes designed to bind to the family of CDKs. The site of crosslinking of the optimised probe, as well as its ability to enrich members of the CDK family from cell lysates, was investigated. In a proof of concept study, we subsequently developed a photoaffinity probe-based competition assay to profile CDK inhibitors. We anticipate that this approach will be widely applicable to the study of small molecule binding to protein families of interest.


Asunto(s)
Marcadores de Afinidad/química , Antineoplásicos/química , Reactivos de Enlaces Cruzados/química , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Isoformas de Proteínas/química , Inhibidores de Proteínas Quinasas/química , Unión Competitiva , Ensayos de Selección de Medicamentos Antitumorales , Espectrometría de Masas , Estructura Molecular , Procesos Fotoquímicos , Roscovitina , Relación Estructura-Actividad
11.
J Am Chem Soc ; 140(3): 932-939, 2018 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-29232121

RESUMEN

Selective covalent inhibition of kinases by targeting poorly conserved cysteines has proven highly fruitful to date in the development of chemical probes and approved drugs. However, this approach is limited to ∼200 kinases possessing such a cysteine near the ATP-binding pocket. Herein, we report a novel approach to achieve selective, irreversible kinase inhibition, by targeting the conserved catalytic lysine residue. We have illustrated our approach by developing selective, covalent PI3Kδ inhibitors that exhibit nanomolar potency in cellular assays, and a duration of action >48 h in CD4+ T cells. Despite conservation of the lysine residue throughout the kinome, the lead compound shows high levels of selectivity over a selection of lipid and protein kinases in biochemical assays, as well as covalent binding to very few off-target proteins in live-cell proteomic studies. We anticipate this approach could offer a general strategy, as an alternative to targeting non-conserved cysteines, for the development of selective covalent kinase inhibitors.


Asunto(s)
Lisina/química , Fosfatidilinositol 3-Quinasas/química , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Animales , Dominio Catalítico/efectos de los fármacos , Línea Celular , Fosfatidilinositol 3-Quinasa Clase I , Descubrimiento de Drogas , Humanos , Lisina/metabolismo , Ratones , Simulación del Acoplamiento Molecular , Fosfatidilinositol 3-Quinasas/metabolismo , Proteómica
12.
Chem Sci ; 4(8): 3110-3117, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26682036

RESUMEN

2-Oxoglutarate and iron dependent oxygenases are therapeutic targets for human diseases. Using a representative 2OG oxygenase panel, we compare the inhibitory activities of 5-carboxy-8-hydroxyquinoline (IOX1) and 4-carboxy-8-hydroxyquinoline (4C8HQ) with that of two other commonly used 2OG oxygenase inhibitors, N-oxalylglycine (NOG) and 2,4-pyridinedicarboxylic acid (2,4-PDCA). The results reveal that IOX1 has a broad spectrum of activity, as demonstrated by the inhibition of transcription factor hydroxylases, representatives of all 2OG dependent histone demethylase subfamilies, nucleic acid demethylases and γ-butyrobetaine hydroxylase. Cellular assays show that, unlike NOG and 2,4-PDCA, IOX1 is active against both cytosolic and nuclear 2OG oxygenases without ester derivatisation. Unexpectedly, crystallographic studies on these oxygenases demonstrate that IOX1, but not 4C8HQ, can cause translocation of the active site metal, revealing a rare example of protein ligand-induced metal movement.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA