Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Curr Drug Metab ; 24(2): 124-130, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36748817

RESUMEN

BACKGROUND: Cytochrome P450 (CYP) 46A1 enzyme is a neuro-specific metabolic enzyme that converts cholesterol to 24-hydroxycholesterol. Inhibition of CYP46A1 activity is of great significance to improve neurodegenerative disorder. OBJECTIVE: The present study aimed to investigate the inhibitory effect of wolfberry dicaffeoylspermidine derivatives on CYP46A1. METHODS: The inhibitory effect of six wolfberry dicaffeoylspermidine derivatives on CYP46A1 activity was investigated using cholesterol as a substrate in vitro. Molecular docking was used to simulate the interactions between wolfberry dicaffeoylspermidine derivatives and CYP46A1. RESULTS: Of these spermidines, lycibarbarspermidines D (1) and A (2) showed highly-selective and strong inhibitory effects on CYP46A1 but not on other human CYP isoforms. Both 1 and 2 exhibit mixed partial competitive inhibition of CYP46A1, with Ki values of 106 nM and 258 nM, respectively. Notably, 1 and 2 had excellent orientations within the active cavity of CYP46A1, and both formed three water-hydrogen bonds with W732 and W765, located near the heme of CYP46A1. CONCLUSION: Compounds 1 and 2 showed a highly-selective and nanomolar affinity for CYP46A1 in vitro. These findings suggested that compounds 1 and 2 could be used as potent inhibitors of CYP46A1 in vitro.


Asunto(s)
Lycium , Humanos , Colesterol 24-Hidroxilasa/química , Colesterol 24-Hidroxilasa/metabolismo , Lycium/metabolismo , Simulación del Acoplamiento Molecular , Colesterol/metabolismo , Sistema Enzimático del Citocromo P-450
2.
Drug Metab Dispos ; 50(7): 923-930, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35489779

RESUMEN

(S)-Efavirenz (EFV) is a reverse transcriptase inhibitor and an antiviral drug. In addition, (S)-EFV can interact off target with CYP46A1, the major cholesterol hydroxylating enzyme in the mammalian brain, and allosterically activate CYP46A1 at a small dose in mice and humans. Studies with purified CYP46A1 identified two allosteric sites on the enzyme surface, one for (S)-EFV and the second site for L-glutamate (Glu), a neurotransmitter that also activates CYP46A1 either alone or in the presence of (S)-EFV. Previously, we found that racemic (rac)-7-hydroxyefavirenz, (rac)-8-hydroxyefavirenz, (S)-8-hydroxyefavirenz, and (rac)-8,14-dihydroxyefavirenz, compounds with the hydroxylation positions corresponding to the metabolism of (S)-EFV in the liver, activated CYP46A1 in vitro. Yet, these compounds differed from (S)-EFV in how they allosterically interacted with CYP46A1. Herein, we further characterized (rac)-7-hydroxyefavirenz, (rac)-8-hydroxyefavirenz, (S)-8-hydroxyefavirenz, and (rac)-8,14-dihydroxyefavirenz, and, in addition, (R)-EFV, (S)-7-hydroxyefavirenz, (rac)-7,8-dihydroxyefavirenz, (S)-7,8-dihydroxyefavirenz, and (S)-8,14-dihydroxyefavirenz for activation and binding to CYP46A1 in vitro. We found that the spatial configuration of all tested compounds neither affected the CYP46A1 activation nor the sites of binding to CYP46A1. Yet, the hydroxylation position determined whether the hydroxylated metabolite interacted with the allosteric site for (S)-EFV [(R)-EFV, (rac)-7,8-dihydroxyefavirenz, and (S)-7,8-dihydroxyefavirenz], L-Glu [(rac)- and (S)-8,14-dihydroxyefavirenz], or both [(rac)-7-hydroxyefavirenz, (S)-7-hydroxyefavirenz, (rac)-8-hydroxyefavirenz, and (S)-8-hydroxyefavirenz]. This difference in binding to the allosteric sites determined, in turn, how CYP46A1 activity was changed in the coincubations with (S)-EFV and either its metabolite or L-Glu. The results suggest EFV metabolites that could be more potent for CYP46A1 activation in vivo than (S)-EFV. SIGNIFICANCE STATEMENT: This study found that not only efavirenz but also all its hydroxylated metabolites allosterically activate CYP46A1 in vitro. The enzyme activation depended on the hydroxylation position but not the metabolite spatial configuration and involved either one or two allosteric sites-for efavirenz, L-glutamate, or both. The results suggest that the hydroxylated efavirenz metabolites may differ from efavirenz in how they interact with the CYP46A1 allosteric and active sites.


Asunto(s)
Benzoxazinas , Colesterol 24-Hidroxilasa , Ácido Glutámico , Alquinos , Animales , Benzoxazinas/química , Colesterol 24-Hidroxilasa/química , Colesterol 24-Hidroxilasa/metabolismo , Ciclopropanos , Ácido Glutámico/metabolismo , Hidroxilación , Ratones
3.
ACS Chem Neurosci ; 13(10): 1526-1533, 2022 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-35438962

RESUMEN

CYP46A1 is an important potential target for the treatment of Alzheimer's disease (AD), which is the most common neurodegenerative disease among older individuals. However, the binding mechanism between CYP46A1 and substrate cholesterol (CH) has not been clarified and will not be conducive to the research of relevant drug molecules. In this study, we integrated molecular docking, molecular dynamics (MD) simulations, and adaptive steered MD simulations to explore the recognition and binding mechanism of CYP46A1 with CH. Two key factors affecting the interaction between CH and CYP46A1 are determined: one is a hydrophobic cavity formed by seven hydrophobic residues (F80, Y109, L112, I222, W368, F371, and T475), which provides nonpolar interactions to stabilize CH, and the other is a hydrogen bond formed by H81 and CH, which ensures the binding direction of CH. In addition, the tunnel analysis results show that tunnel 2a is identified as the primary pathway of CH. The entry of CH induces tunnel 2e to close and tunnel w to open. Our results may provide effective clues for the design of drugs based on the substrate for AD and improve our understanding of the structure-function of CYP46A1.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Colesterol/metabolismo , Colesterol 24-Hidroxilasa/química , Colesterol 24-Hidroxilasa/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Reconocimiento en Psicología
4.
J Med Chem ; 63(12): 6477-6488, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31617715

RESUMEN

Cytochrome P450 46A1 (CYP46A1) is a central nervous system-specific enzyme, which catalyzes cholesterol 24-hydroxylation. Currently CYP46A1 is being evaluated in a clinical trial for activation by small doses of the anti-HIV drug efavirenz. Eight efavirenz-related compounds were investigated for CYP46A1 activation in vitro, induction of a CYP46A1 spectral response, spectral Kd values, interaction with the P450 allosteric sites, and a model of binding to the enzyme active site. We gained insight into structure-activity relationships of efavirenz for CYP46A1 activation and found that the investigated efavirenz primary metabolites are stronger and better activators of CYP46A1 than efavirenz. We also established that CYP46A1 is activated by racemates and that a conformational-selection mechanism is operative in CYP46A1. The results suggest structural modifications of efavirenz to further increase CYP46A1 activation without inhibition at high compound concentrations. It is possible that not only efavirenz but its metabolites activate CYP46A1 in vivo.


Asunto(s)
Benzoxazinas/química , Benzoxazinas/farmacología , Colesterol 24-Hidroxilasa/metabolismo , Inductores del Citocromo P-450 CYP3A/química , Inductores del Citocromo P-450 CYP3A/farmacología , Preparaciones Farmacéuticas/metabolismo , Alquinos , Sitio Alostérico , Dominio Catalítico , Colesterol/metabolismo , Colesterol 24-Hidroxilasa/química , Ciclopropanos , Humanos , Hidroxilación , Técnicas In Vitro , Preparaciones Farmacéuticas/química , Conformación Proteica
5.
J Med Chem ; 63(12): 6474-6476, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31845585

RESUMEN

Cytochrome P450 46A1 (CYP46A1) or cholesterol-24-hydroxylase is responsible for cholesterol metabolism and homeostasis in the human brain. More recently its activity has been linked to brain function and disease. The anti-HIV drug efavirenz activates CYP46A1 at low drug levels while inhibiting the enzyme activity at the high dose used in clinical practice. Synthetic analogs and hydroxylated metabolites of efavirenz enhance CYP46A1 activity, with reduced unwanted enzyme inhibition at higher concentrations. These observations provide a platform for structural modifications of efavirenz to modulate CYP46A1 activity as a therapeutic target of brain disorders such as Alzheimer's disease, for which currently no treatment is available.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Benzoxazinas/uso terapéutico , Encéfalo/efectos de los fármacos , Colesterol 24-Hidroxilasa/metabolismo , Inductores del Citocromo P-450 CYP3A/uso terapéutico , Alquinos , Enfermedad de Alzheimer/patología , Animales , Benzoxazinas/química , Colesterol 24-Hidroxilasa/química , Ciclopropanos , Inductores del Citocromo P-450 CYP3A/química , Humanos
6.
J Biol Chem ; 292(31): 12934-12946, 2017 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-28642370

RESUMEN

Cytochrome P450 46A1 (CYP46A1, cholesterol 24-hydroxylase) is the enzyme responsible for the majority of cholesterol elimination from the brain. Previously, we found that the anti-HIV drug efavirenz (EFV) can pharmacologically activate CYP46A1 in mice. Herein, we investigated whether CYP46A1 could also be activated by endogenous compounds, including major neurotransmitters. In vitro experiments with purified recombinant CYP46A1 indicated that CYP46A1 is activated by l-glutamate (l-Glu), l-aspartate, γ-aminobutyric acid, and acetylcholine, with l-Glu eliciting the highest increase (3-fold) in CYP46A1-mediated cholesterol 24-hydroxylation. We also found that l-Glu and other activating neurotransmitters bind to the same site on the CYP46A1 surface, which differs from the EFV-binding site. The other principal differences between EFV and l-Glu in CYP46A1 activation include an apparent lack of l-Glu binding to the P450 active site and different pathways of signal transduction from the allosteric site to the active site. EFV and l-Glu similarly increased the CYP46A1 kcat, the rate of the "fast" phase of the enzyme reduction by the redox partner NADPH-cytochrome P450 oxidoreductase, and the amount of P450 reduced. Spectral titrations with cholesterol, in the presence of EFV or l-Glu, suggest that water displacement from the heme iron can be affected in activator-bound CYP46A1. Moreover, EFV and l-Glu synergistically activated CYP46A1. Collectively, our in vitro data, along with those from previous cell culture and in vivo studies by others, suggest that l-Glu-induced CYP46A1 activation is of physiological relevance.


Asunto(s)
Acetilcolina/metabolismo , Ácido Aspártico/metabolismo , Colesterol 24-Hidroxilasa/metabolismo , Ácido Glutámico/metabolismo , Modelos Moleculares , Proteínas del Tejido Nervioso/agonistas , Ácido gamma-Aminobutírico/metabolismo , Acetilcolina/química , Alquinos , Regulación Alostérica/efectos de los fármacos , Sustitución de Aminoácidos , Fármacos Anti-VIH/química , Fármacos Anti-VIH/metabolismo , Fármacos Anti-VIH/farmacología , Ácido Aspártico/química , Benzoxazinas/química , Benzoxazinas/metabolismo , Benzoxazinas/farmacología , Sitios de Unión , Biocatálisis/efectos de los fármacos , Colesterol 24-Hidroxilasa/química , Colesterol 24-Hidroxilasa/genética , Ciclopropanos , Medición de Intercambio de Deuterio , Activación Enzimática/efectos de los fármacos , Ácido Glutámico/química , Ligandos , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Mutación , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Conformación Proteica , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Ácido gamma-Aminobutírico/química
7.
J Biol Chem ; 291(22): 11876-86, 2016 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-27056331

RESUMEN

Cytochrome P450 46A1 (CYP46A1) is a microsomal enzyme and cholesterol 24-hydroxylase that controls cholesterol elimination from the brain. This P450 is also a potential target for Alzheimer disease because it can be activated pharmacologically by some marketed drugs, as exemplified by efavirenz, the anti-HIV medication. Previously, we suggested that pharmaceuticals activate CYP46A1 allosterically through binding to a site on the cytosolic protein surface, which is different from the enzyme active site facing the membrane. Here we identified this allosteric site for efavirenz on CYP46A1 by using a combination of hydrogen-deuterium exchange coupled to MS, computational modeling, site-directed mutagenesis, and analysis of the CYP46A1 crystal structure. We also mapped the binding region for the CYP46A1 redox partner oxidoreductase and found that the allosteric and redox partner binding sites share a common border. On the basis of the data obtained, we propose the mechanism of CYP46A1 allostery and the pathway for the signal transmission from the P450 allosteric site to the active site.


Asunto(s)
Benzoxazinas/farmacología , Colesterol 24-Hidroxilasa/metabolismo , Colesterol/metabolismo , Inhibidores de la Transcriptasa Inversa/farmacología , Alquinos , Sitio Alostérico , Secuencia de Aminoácidos , Sitios de Unión , Dominio Catalítico , Colesterol 24-Hidroxilasa/química , Colesterol 24-Hidroxilasa/genética , Cristalografía por Rayos X , Ciclopropanos , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación/genética , Unión Proteica , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA