Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38105987

RESUMEN

SIRT5 is a sirtuin deacylase that represents the major activity responsible for removal of negatively-charged lysine modifications, in the mitochondrial matrix and elsewhere in the cell. In benign cells and mouse models, under basal non-stressed conditions, the phenotypes of SIRT5 deficiency are generally quite subtle. Here, we identify two homozygous SIRT5 variants in human patients suffering from severe mitochondrial disease. Both variants, P114T and L128V, are associated with reduced SIRT5 protein stability and impaired biochemical activity, with no evidence of neomorphic or dominant negative properties. The crystal structure of the P114T enzyme was solved and shows only subtle deviations from wild-type. Via CRISPR-Cas9, we generate a mouse model that recapitulates the human P114T mutation; homozygotes show reduced SIRT5 levels and activity, but no obvious metabolic abnormalities, neuropathology or other gross evidence of severe disease. We conclude that these human SIRT5 variants most likely represent severe hypomorphs, and are likely not the primary pathogenic cause of the neuropathology observed in the patients.

2.
J Biol Chem ; 299(3): 102999, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36773804

RESUMEN

Abiraterone acetate is a first-line therapy for castration-resistant prostate cancer. This prodrug is deacetylated in vivo to abiraterone, which is a potent and specific inhibitor of cytochrome P450 17A1 (CYP17A1). CYP17A1 performs two sequential steps that are required for the biosynthesis of androgens that drive prostate cancer proliferation, analogous to estrogens in breast cancer. Abiraterone can be further metabolized in vivo on the steroid A ring to multiple metabolites that also inhibit CYP17A1. Despite its design as an active-site-directed substrate analog, abiraterone and its metabolites demonstrate mixed competitive/noncompetitive inhibition. To understand their binding, we solved the X-ray structures of CYP17A1 with three primary abiraterone metabolites. Despite different conformations of the steroid A ring and substituents, all three bound in the CYP17A1 active site with the steroid core packed against the I helix and the A ring C3 keto or hydroxyl oxygen forming a hydrogen bond with N202 similar to abiraterone itself. The structure of CYP17A1 with 3-keto, 5α-abiraterone was solved to 2.0 Å, the highest resolution to date for a CYP17A1 complex. This structure had additional electron density near the F/G loop, which is likely a second molecule of the inhibitor and which may explain the noncompetitive inhibition. Mutation of the adjacent Asn52 to Tyr positions its side chain in this space, maintains enzyme activity, and prevents binding of the peripheral ligand. Collectively, our findings provide further insight into abiraterone metabolite binding and CYP17A1 function.


Asunto(s)
Antineoplásicos , Neoplasias de la Próstata , Masculino , Humanos , Antineoplásicos/química , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Sitios de Unión , Esteroides/química , Sistema Enzimático del Citocromo P-450/metabolismo , Esteroide 17-alfa-Hidroxilasa/metabolismo
3.
J Med Chem ; 64(19): 14540-14556, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34613724

RESUMEN

Embryonic ectoderm development (EED) is a promising therapeutic target for human cancers and other diseases. We report herein the discovery of exceptionally potent and efficacious EED inhibitors. By conformational restriction of a previously reported EED inhibitor, we obtained a potent lead compound. Further optimization of the lead yielded exceptionally potent EED inhibitors. The best compound EEDi-5273 binds to EED with an IC50 value of 0.2 nM and inhibits the KARPAS422 cell growth with an IC50 value of 1.2 nM. It demonstrates an excellent PK and ADME profile, and its oral administration leads to complete and persistent tumor regression in the KARPAS422 xenograft model with no signs of toxicity. Co-crystal structures of two potent EED inhibitors with EED provide a solid structural basis for their high-affinity binding. EEDi-5273 is a promising EED inhibitor for further advanced preclinical development for the treatment of human cancer and other human diseases.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Administración Oral , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Humanos , Neoplasias/patología , Relación Estructura-Actividad
4.
ACS Med Chem Lett ; 11(6): 1348-1352, 2020 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-32551023

RESUMEN

The mixed-lineage leukemia (MLL) protein, also known as MLL1, is a lysine methyltransferase specifically responsible for methylation of histone 3 lysine 4. MLL has been pursued as an attractive therapeutic target for the treatment of acute leukemia carrying the MLL fusion gene or MLL leukemia. Herein, we report the design, synthesis, and evaluation of an S-adenosylmethionine-based focused chemical library which led to the discovery of potent small-molecule inhibitors directly targeting the MLL SET domain. Determination of cocrystal structures for a number of these MLL inhibitors reveals that they adopt a unique binding mode that locks the MLL SET domain in an open, inactive conformation.

5.
J Med Chem ; 63(13): 7252-7267, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32580550

RESUMEN

Inhibition of embryonic ectoderm development (EED) is a new cancer therapeutic strategy. Herein, we report our discovery of EEDi-5285 as an exceptionally potent, efficacious, and orally active EED inhibitor. EEDi-5285 binds to the EED protein with an IC50 value of 0.2 nM and inhibits cell growth with IC50 values of 20 pM and 0.5 nM in the Pfeiffer and KARPAS422 lymphoma cell lines, respectively, carrying an EZH2 mutation. EEDi-5285 is approximately 100 times more potent than EED226 in binding to EED and >300 times more potent than EED226 in inhibition of cell growth in the KARPAS422 cell line. EEDi-5285 has excellent pharmacokinetics and achieves complete and durable tumor regression in the KARPAS422 xenograft model in mice with oral administration. The cocrystal structure of EEDi-5285 in a complex with EED defines the precise structural basis for their high binding affinity. EEDi-5285 is the most potent and efficacious EED inhibitor reported to date.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Complejo Represivo Polycomb 2/química , Bibliotecas de Moléculas Pequeñas/farmacología , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Disponibilidad Biológica , Línea Celular Tumoral , Cristalografía por Rayos X , Histonas/metabolismo , Humanos , Linfoma/tratamiento farmacológico , Linfoma/patología , Ratones SCID , Complejo Represivo Polycomb 2/metabolismo , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Med Chem ; 62(6): 3068-3087, 2019 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-30735370

RESUMEN

Using reported glutathione S-transferase omega 1 (GSTO1-1) cocrystal structures, we designed and synthesized acrylamide-containing compounds that covalently bind to Cys32 on the catalytic site. Starting from a thiazole derivative 10 (GSTO1-1 IC50 = 0.6 µM), compound 18 was synthesized and cocrystallized with GSTO1. Modification on the amide moiety of hit compound 10 significantly increased the GSTO1-1 inhibitory potency. We solved the cocrystal structures of new derivatives, 37 and 44, bearing an amide side chain bound to GSTO1. These new structures showed a reorientation of the phenyl thiazole core of inhibitors, 37 and 44, when compared to 18. Guided by the cocrystal structure of GSTO1:44, analogue 49 was designed, resulting in the most potent GSTO1-1 inhibitor (IC50 = 0.22 ± 0.02 nM) known to date. We believe that our data will form the basis for future studies of developing GSTO1-1 as a new drug target for cancer therapy.


Asunto(s)
Acrilamidas/química , Acrilamidas/farmacología , Diseño de Fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Glutatión Transferasa/antagonistas & inhibidores , Acrilamidas/farmacocinética , Animales , Dominio Catalítico , Cristalografía por Rayos X , Células HCT116 , Semivida , Humanos , Ratones , Microsomas Hepáticos/enzimología , Microsomas Hepáticos/metabolismo , Estructura Molecular , Relación Estructura-Actividad
7.
J Med Chem ; 61(12): 5162-5186, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29727569

RESUMEN

We pursued a structure-guided approach toward the development of improved dihydroorotate dehydrogenase (DHODH) inhibitors with the goal of forming new interactions between DHODH and the brequinar class of inhibitors. Two potential residues, T63 and Y356, suitable for novel H-bonding interactions, were identified in the brequinar-binding pocket. Analogues were designed to maintain the essential pharmacophore and form new electrostatic interactions through strategically positioned H-bond accepting groups. This effort led to the discovery of potent quinoline-based analogues 41 (DHODH IC50 = 9.71 ± 1.4 nM) and 43 (DHODH IC50 = 26.2 ± 1.8 nM). A cocrystal structure between 43 and DHODH depicts a novel water mediated H-bond interaction with T63. Additional optimization led to the 1,7-naphthyridine 46 (DHODH IC50 = 28.3 ± 3.3 nM) that forms a novel H-bond with Y356. Importantly, compound 41 possesses significant oral bioavailability ( F = 56%) and an elimination t1/2 = 2.78 h (PO dosing). In conclusion, the data supports further preclinical studies of our lead compounds toward selection of a candidate for early-stage clinical development.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/antagonistas & inhibidores , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/química , Quinolinas/química , Administración Oral , Animales , Compuestos de Bifenilo/química , Cristalografía por Rayos X , Dihidroorotato Deshidrogenasa , Diseño de Fármacos , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/síntesis química , Femenino , Células HCT116 , Semivida , Humanos , Enlace de Hidrógeno , Ratones Endogámicos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Piridinas/química , Pirimidinas/química , Solubilidad , Relación Estructura-Actividad , Termodinámica
8.
Drug Metab Dispos ; 45(6): 635-645, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28373265

RESUMEN

Human steroidogenic cytochrome P450 17A1 (CYP17A1) is a bifunctional enzyme that performs both hydroxylation and lyase reactions, with the latter required to generate androgens that fuel prostate cancer proliferation. The steroid abiraterone, the active form of the only CYP17A1 inhibitor approved by the Food and Drug Administration, binds the catalytic heme iron, nonselectively impeding both reactions and ultimately causing undesirable corticosteroid imbalance. Some nonsteroidal inhibitors reportedly inhibit the lyase reaction more than the preceding hydroxylase reaction, which would be clinically advantageous, but the mechanism is not understood. Thus, the nonsteroidal inhibitors seviteronel and orteronel and the steroidal inhibitors abiraterone and galeterone were compared with respect to their binding modes and hydroxylase versus lyase inhibition. Binding studies and X-ray structures of CYP17A1 with nonsteroidal inhibitors reveal coordination to the heme iron like the steroidal inhibitors. (S)-seviteronel binds similarly to both observed CYP17A1 conformations. However, (S)-orteronel and (R)-orteronel bind to distinct CYP17A1 conformations that differ in a region implicated in ligand entry/exit and the presence of a peripheral ligand. To reconcile these binding modes with enzyme function, side-by-side enzymatic analysis was undertaken and revealed that neither the nonsteroidal seviteronel nor the (S)-orteronel inhibitors demonstrated significant lyase selectivity, but the less potent (R)-orteronel was 8- to 11-fold selective for lyase inhibition. While active-site iron coordination is consistent with competitive inhibition, conformational selection for binding of some inhibitors and the differential presence of a peripheral ligand molecule suggest the possibility of CYP17A1 functional modulation by features outside the active site.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Esteroide 17-alfa-Hidroxilasa/antagonistas & inhibidores , Esteroides/química , Esteroides/farmacología , Androstadienos/química , Androstadienos/farmacología , Androstenos/química , Androstenos/farmacología , Bencimidazoles/química , Bencimidazoles/farmacología , Dominio Catalítico/efectos de los fármacos , Humanos , Imidazoles/química , Imidazoles/farmacología , Naftalenos/química , Naftalenos/farmacología
9.
J Biol Chem ; 292(17): 7173-7188, 2017 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-28228478

RESUMEN

The transforming growth factor ß isoforms, TGF-ß1, -ß2, and -ß3, are small secreted homodimeric signaling proteins with essential roles in regulating the adaptive immune system and maintaining the extracellular matrix. However, dysregulation of the TGF-ß pathway is responsible for promoting the progression of several human diseases, including cancer and fibrosis. Despite the known importance of TGF-ßs in promoting disease progression, no inhibitors have been approved for use in humans. Herein, we describe an engineered TGF-ß monomer, lacking the heel helix, a structural motif essential for binding the TGF-ß type I receptor (TßRI) but dispensable for binding the other receptor required for TGF-ß signaling, the TGF-ß type II receptor (TßRII), as an alternative therapeutic modality for blocking TGF-ß signaling in humans. As shown through binding studies and crystallography, the engineered monomer retained the same overall structure of native TGF-ß monomers and bound TßRII in an identical manner. Cell-based luciferase assays showed that the engineered monomer functioned as a dominant negative to inhibit TGF-ß signaling with a Ki of 20-70 nm Investigation of the mechanism showed that the high affinity of the engineered monomer for TßRII, coupled with its reduced ability to non-covalently dimerize and its inability to bind and recruit TßRI, enabled it to bind endogenous TßRII but prevented it from binding and recruiting TßRI to form a signaling complex. Such engineered monomers provide a new avenue to probe and manipulate TGF-ß signaling and may inform similar modifications of other TGF-ß family members.


Asunto(s)
Ingeniería de Proteínas/métodos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Transducción de Señal , Factor de Crecimiento Transformador beta/química , Secuencias de Aminoácidos , Animales , Progresión de la Enfermedad , Matriz Extracelular/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Humanos , Cinética , Ratones , Unión Proteica , Pliegue de Proteína , Isoformas de Proteínas , Multimerización de Proteína , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Solubilidad , Resonancia por Plasmón de Superficie , Factor de Crecimiento Transformador beta/metabolismo , Ultracentrifugación
10.
Mol Cell Endocrinol ; 441: 68-75, 2017 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-27566228

RESUMEN

Cytochrome P450 17A1 (CYP17A1) operates at the core of human steroidogenesis, directing precursors into mineralocorticoids, glucocorticoids, or sex steroids. Although the 17α-hydroxylase and 17,20-lyase activities of this dual function enzyme have been investigated extensively, until recently no CYP17A1 structures were available to inform our understanding. Structures of CYP17A1 with a range of steroidal inhibitors and substrates are now available. This review relates functional knowledge of this enzyme to structural features defining the selective differentiation between its various substrates. While both hydroxylase and lyase substrates have similar orientations with respect to the heme, subtle differences in hydrogen bonding between CYP17A1 and the C3 substituent at the opposite end of ligands appear to correlate with differential substrate utilization and product formation. Complementary structural information from solution NMR supports cytochrome b5 allosteric modulation of the lyase reaction, implicating regions involved in ligand access to the otherwise buried active site.


Asunto(s)
Esteroide 17-alfa-Hidroxilasa/química , Esteroide 17-alfa-Hidroxilasa/metabolismo , Animales , Humanos , Hidroxilación , Pregnenolona/metabolismo , Progesterona/metabolismo , Estereoisomerismo , Especificidad por Sustrato
11.
Sci Rep ; 6: 29468, 2016 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-27406023

RESUMEN

Cytochrome P450 17A1 (CYP17A1) is an important target in the treatment of prostate cancer because it produces androgens required for tumour growth. The FDA has approved only one CYP17A1 inhibitor, abiraterone, which contains a steroidal scaffold similar to the endogenous CYP17A1 substrates. Abiraterone is structurally similar to the substrates of other cytochrome P450 enzymes involved in steroidogenesis, and interference can pose a liability in terms of side effects. Using non-steroidal scaffolds is expected to enable the design of compounds that interact more selectively with CYP17A1. Therefore, we combined a structure-based virtual screening approach with density functional theory (DFT) calculations to suggest non-steroidal compounds selective for CYP17A1. In vitro assays demonstrated that two such compounds selectively inhibited CYP17A1 17α-hydroxylase and 17,20-lyase activities with IC50 values in the nanomolar range, without affinity for the major drug-metabolizing CYP2D6 and CYP3A4 enzymes and CYP21A2, with the latter result confirmed in human H295R cells.


Asunto(s)
Diseño de Fármacos , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/tratamiento farmacológico , Esteroide 17-alfa-Hidroxilasa/antagonistas & inhibidores , Androstenos/farmacología , Dominio Catalítico , Línea Celular Tumoral , Supervivencia Celular , Cromatografía Liquida , Cristalografía por Rayos X , Humanos , Concentración 50 Inhibidora , Ligandos , Masculino , Espectrometría de Masas , Simulación del Acoplamiento Molecular , Nitrógeno/química , Próstata/metabolismo , Unión Proteica , Esteroide 17-alfa-Hidroxilasa/sangre , Esteroide 17-alfa-Hidroxilasa/metabolismo , Esteroides
12.
J Biol Chem ; 289(47): 32952-64, 2014 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-25301938

RESUMEN

The human cytochrome P450 17A1 (CYP17A1) enzyme operates at a key juncture of human steroidogenesis, controlling the levels of mineralocorticoids influencing blood pressure, glucocorticoids involved in immune and stress responses, and androgens and estrogens involved in development and homeostasis of reproductive tissues. Understanding CYP17A1 multifunctional biochemistry is thus integral to treating prostate and breast cancer, subfertility, blood pressure, and other diseases. CYP17A1 structures with all four physiologically relevant steroid substrates suggest answers to four fundamental aspects of CYP17A1 function. First, all substrates bind in a similar overall orientation, rising ∼60° with respect to the heme. Second, both hydroxylase substrates pregnenolone and progesterone hydrogen bond to Asn(202) in orientations consistent with production of 17α-hydroxy major metabolites, but functional and structural evidence for an A105L mutation suggests that a minor conformation may yield the minor 16α-hydroxyprogesterone metabolite. Third, substrate specificity of the subsequent 17,20-lyase reaction may be explained by variation in substrate height above the heme. Although 17α-hydroxyprogesterone is only observed farther from the catalytic iron, 17α-hydroxypregnenolone is also observed closer to the heme. In conjunction with spectroscopic evidence, this suggests that only 17α-hydroxypregnenolone approaches and interacts with the proximal oxygen of the catalytic iron-peroxy intermediate, yielding efficient production of dehydroepiandrosterone as the key intermediate in human testosterone and estrogen synthesis. Fourth, differential positioning of 17α-hydroxypregnenolone offers a mechanism whereby allosteric binding of cytochrome b5 might selectively enhance the lyase reaction. In aggregate, these structures provide a structural basis for understanding multiple key reactions at the heart of human steroidogenesis.


Asunto(s)
Dominio Catalítico , Estructura Secundaria de Proteína , Esteroide 17-alfa-Hidroxilasa/química , Esteroide 17-alfa-Hidroxilasa/metabolismo , 17-alfa-Hidroxiprogesterona/química , 17-alfa-Hidroxiprogesterona/metabolismo , Androstenos , Androstenoles/química , Androstenoles/metabolismo , Sitios de Unión/genética , Cristalografía por Rayos X , Deshidroepiandrosterona/química , Deshidroepiandrosterona/metabolismo , Estrógenos/metabolismo , Hemo/metabolismo , Humanos , Enlace de Hidrógeno , Cinética , Modelos Moleculares , Estructura Molecular , Mutación , Oxidación-Reducción , Pregnenolona/química , Pregnenolona/metabolismo , Progesterona/química , Progesterona/metabolismo , Unión Proteica , Esteroide 17-alfa-Hidroxilasa/genética , Esteroides/química , Esteroides/metabolismo , Especificidad por Sustrato , Testosterona/metabolismo
13.
Phytochemistry ; 72(7): 662-7, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21338993

RESUMEN

An acylphloroglucinol, elliptophenone A, and two xanthones, elliptoxanthone A and elliptoxanthone B, were isolated from the aerial portions of Hypericum ellipticum together with three known xanthones, 1,3,7-trihydroxy-8-(3-methyl-2-butenyl)-9H-xanthen-9-one, 1,6-dihydroxy-4-methoxy-9H-xanthen-9-one, and 1,4,5-trihydroxy-9H-xanthen-9-one. Their structures were determined by spectroscopic analyses. The acylphloroglucinol and xanthones were evaluated for cytotoxicity using three human colon cancer cell lines cell lines (HT-29, HCT-116 and Caco-2) and a normal human colon cell line (CCD-18Co).


Asunto(s)
Hypericum/química , Floroglucinol/análogos & derivados , Xantonas/química , Xantonas/farmacología , Línea Celular Tumoral , Humanos , Concentración 50 Inhibidora , Floroglucinol/química , Floroglucinol/aislamiento & purificación , Floroglucinol/farmacología , Xantonas/aislamiento & purificación
14.
Nat Prod Commun ; 4(4): 507-10, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19475994

RESUMEN

Examination of the acetone extract of the aerial parts of Hypericum ellipticum afforded a new acetylated benzophenone glucoside (3'-O-beta-D-3",4",6"-triacetylglucopyranosyl-2,4,5',6-tetrahydroxybenzophenone) together with catechin and epicatechin. The structure of the benzophenone glucoside was determined by 2D NMR spectroscopic data. The compound inhibited the proliferation of CNS tumor cell line (SF-268) and lipid peroxidation in in vitro assays.


Asunto(s)
Benzofenonas/química , Citostáticos/química , Glucósidos/química , Hypericum/química , Benzofenonas/aislamiento & purificación , Benzofenonas/farmacología , Catequina/química , Catequina/aislamiento & purificación , Catequina/farmacología , Línea Celular Tumoral , Citostáticos/aislamiento & purificación , Citostáticos/farmacología , Glucósidos/aislamiento & purificación , Glucósidos/farmacología , Humanos , Peroxidación de Lípido/efectos de los fármacos , Resonancia Magnética Nuclear Biomolecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...