Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 315
Filtrar
1.
Biochem Pharmacol ; 194: 114824, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34748821

RESUMEN

Diphenylamine NSAIDs are taken frequently for chronic pain conditions, yet their use may potentiate hepatotoxicity risks through poorly characterized metabolic mechanisms. Our previous work revealed that seven marketed or withdrawn diphenylamine NSAIDs undergo bioactivation into quinone-species metabolites, whose reaction specificities depended on halogenation and the type of acidic group on the diphenylamine. Herein, we identified cytochromes P450 responsible for those bioactivations, determined reaction specificities, and estimated relative contributions of enzymes to overall hepatic bioactivations and detoxifications. A qualitative activity screen revealed CYP2C8, 2C9, 2C19, and 3A4 played roles in drug bioactivation. Subsequent steady-state studies with recombinant CYPs recapitulated the importance of halogenation and acidic group type on bioactivations but importantly, showed patterns unique to each CYP. CYP2C9, 2C19 and 3A4 bioactivated all NSAIDs with CYP2C9 dominating all possible bioactivation pathways. For each CYP, specificities for overall oxidative metabolism were not impacted significantly by differences in NSAID structures but the values themselves differed among the enzymes such that CYP2C9 and 3A4 were more efficient than others. When considering hepatic CYP abundance, CYP2C9 almost exclusively accounted for diphenylamine NSAID bioactivations, whereas CYP3A4 provided a critical counterbalance favoring their overall detoxification. Preference for either outcome would depend on molecular structures favoring metabolism by the CYPs as well as the influence of clinical factors altering their expression and/or activity. While focused on NSAIDs, these findings have broader implications on bioactivation risks given the expansion of the diphenylamine scaffold to other drug classes such as targeted cancer therapeutics.


Asunto(s)
Antiinflamatorios no Esteroideos/metabolismo , Citocromo P-450 CYP2C9/metabolismo , Citocromo P-450 CYP3A/metabolismo , Difenilamina/metabolismo , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Antiinflamatorios no Esteroideos/toxicidad , Difenilamina/toxicidad , Humanos , Inactivación Metabólica/efectos de los fármacos , Inactivación Metabólica/fisiología , Especificidad por Sustrato/efectos de los fármacos , Especificidad por Sustrato/fisiología
2.
Cell Rep ; 37(7): 110004, 2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34788624

RESUMEN

Polyphosphate (polyP) is a polymer of hundreds of phosphate residues present in all organisms. In mammals, polyP is involved in crucial physiological processes, including coagulation, inflammation, and stress response. However, after decades of research, the metabolic enzymes are still unknown. Here, we purify and identify Nudt3, a NUDIX family member, as the enzyme responsible for polyP phosphatase activity in mammalian cells. We show that Nudt3 shifts its substrate specificity depending on the cation; specifically, Nudt3 is active on polyP when Zn2+ is present. Nudt3 has in vivo polyP phosphatase activity in human cells, and importantly, we show that cells with altered polyP levels by modifying Nudt3 protein amount present reduced viability upon oxidative stress and increased DNA damage, suggesting that polyP and Nudt3 play a role in oxidative stress protection. Finally, we show that Nudt3 is involved in the early stages of embryo development in zebrafish.


Asunto(s)
Ácido Anhídrido Hidrolasas/metabolismo , Estrés Oxidativo/fisiología , Polifosfatos/metabolismo , Ácido Anhídrido Hidrolasas/genética , Ácido Anhídrido Hidrolasas/fisiología , Animales , Células HEK293 , Humanos , Masculino , Mamíferos/metabolismo , Oxidación-Reducción , Monoéster Fosfórico Hidrolasas/fisiología , Ratas , Ratas Sprague-Dawley , Especificidad por Sustrato/fisiología , Pez Cebra , Zinc/metabolismo
3.
Biochem J ; 478(17): 3179-3184, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34492095

RESUMEN

Apoptosis is a cell death program that is executed by the caspases, a family of cysteine proteases that typically cleave after aspartate residues during a proteolytic cascade that systematically dismantles the dying cell. Extensive signaling crosstalk occurs between caspase-mediated proteolysis and kinase-mediated phosphorylation, enabling integration of signals from multiple pathways into the decision to commit to apoptosis. A new study from Maluch et al. examines how phosphorylation within caspase cleavage sites impacts the efficiency of substrate cleavage. The results demonstrate that while phosphorylation in close proximity to the scissile bond is generally inhibitory, it does not necessarily abrogate substrate cleavage, but instead attenuates the rate. In some cases, this inhibition can be overcome by additional favorable substrate features. These findings suggest potential nuanced physiological roles for phosphorylation of caspase substrates with exciting implications for targeting caspases with chemical probes and therapeutics.


Asunto(s)
Caspasas/metabolismo , Fosfotransferasas/metabolismo , Proteolisis , Transducción de Señal/fisiología , Apoptosis/fisiología , Ácido Aspártico/metabolismo , Humanos , Cinética , Fosforilación/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Especificidad por Sustrato/fisiología
4.
Drug Metab Dispos ; 49(3): 254-264, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33376106

RESUMEN

The ability to predict human liver-to-plasma unbound partition coefficient (Kpuu) is important to estimate unbound liver concentration for drugs that are substrates of hepatic organic anion-transporting peptide (OATP) transporters with asymmetric distribution into the liver relative to plasma. Herein, we explored the utility of PXB chimeric mice with humanized liver that are highly repopulated with human hepatocytes to predict human hepatic disposition of OATP substrates, including rosuvastatin, pravastatin, pitavastatin, valsartan, and repaglinide. In vitro total uptake clearance and transporter-mediated active uptake clearance in C57 mouse hepatocytes were greater than in PXB chimeric mouse hepatocytes for rosuvastatin, pravastatin, pitavastatin, and valsartan. Consistent with in vitro uptake data, enhanced hepatic uptake and resulting total systemic clearance were observed with the above four compounds in severely compromised immune-deficient (SCID) control mice compared with the PXB chimeric mice, which suggest that mouse has a stronger transporter-mediated hepatic uptake than human. In vivo liver-to-plasma Kpuu from PXB chimeric and SCID control mice were also compared, and rosuvastatin and pravastatin Kpuu in SCID mice were more than 10-fold higher than that in PXB chimeric mice, whereas pitavastatin, valsartan, and repaglinide Kpuu in SCID mice were comparable with Kpuu in PXB chimeric mice. Finally, PXB chimeric mouse liver-to-plasma Kpuu values were compared with the reported human Kpuu, and a good correlation was observed as the PXB Kpuu vales were within 3-fold of human Kpuu Our results indicate that PXB mice could be a useful tool to delineate hepatic uptake and enable prediction of human liver-to-plasma Kpuu of hepatic uptake transporter substrates. SIGNIFICANCE STATEMENT: We evaluated PXB mouse with humanized liver for its ability to predict human liver disposition of five organic anion-transporting polypeptide transporter substrates. Both in vitro and in vivo data suggest that mouse liver has a stronger transporter-mediated hepatic uptake than the humanized liver in PXB mouse. More importantly, PXB liver-to-plasma unbound partition coefficient (Kpuu) values were compared with the reported human Kpuu, and a good correlation was observed. PXB mice could be a useful tool to project human liver-to-plasma Kpuu of hepatic uptake transporter substrates.


Asunto(s)
Quimera/genética , Quimera/metabolismo , Hígado/metabolismo , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Predicción , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Pravastatina/farmacología , Especificidad por Sustrato/efectos de los fármacos , Especificidad por Sustrato/fisiología
5.
J Cell Physiol ; 236(5): 3220-3233, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33094504

RESUMEN

Protein palmitoylation, in which C16 fatty acid chains are attached to cysteine residues via a reversible thioester linkage, is one of the most common lipid modifications and plays important roles in regulating protein stability, subcellular localization, membrane trafficking, interactions with effector proteins, enzymatic activity, and a variety of other cellular processes. Moreover, the unique reversibility of palmitoylation allows proteins to be rapidly shuttled between biological membranes and cytoplasmic substrates in a process usually controlled by a member of the DHHC family of protein palmitoyl transferases (PATs). Notably, mutations in PATs are closely related to a variety of human diseases, such as cancer, neurological disorders, and immune deficiency conditions. In addition to PATs, intracellular palmitoylation dynamics are also regulated by the interplay between distinct posttranslational modifications, including ubiquitination and phosphorylation. Understanding the specific mechanisms of palmitoylation may reveal novel potential therapeutic targets for many human diseases.


Asunto(s)
Aciltransferasas/metabolismo , Lipoilación/genética , Proteínas de la Membrana/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Animales , Metilación de ADN/fisiología , Humanos , Especificidad por Sustrato/fisiología
6.
Proc Natl Acad Sci U S A ; 117(47): 29609-29617, 2020 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-33168729

RESUMEN

P-glycoprotein (P-gp), also known as ABCB1, is a cell membrane transporter that mediates the efflux of chemically dissimilar amphipathic drugs and confers resistance to chemotherapy in most cancers. Homologous transmembrane helices (TMHs) 6 and 12 of human P-gp connect the transmembrane domains with its nucleotide-binding domains, and several residues in these TMHs contribute to the drug-binding pocket. To investigate the role of these helices in the transport function of P-gp, we substituted a group of 14 conserved residues (seven in both TMHs 6 and 12) with alanine and generated a mutant termed 14A. Although the 14A mutant lost the ability to pump most of the substrates tested out of cancer cells, surprisingly, it acquired a new function. It was able to import four substrates, including rhodamine 123 (Rh123) and the taxol derivative flutax-1. Similar to the efflux function of wild-type P-gp, we found that uptake by the 14A mutant is ATP hydrolysis-, substrate concentration-, and time-dependent. Consistent with the uptake function, the mutant P-gp also hypersensitizes HeLa cells to Rh123 by 2- to 2.5-fold. Further mutagenesis identified residues from both TMHs 6 and 12 that synergistically form a switch in the central region of the two helices that governs whether a given substrate is pumped out of or into the cell. Transforming P-gp or an ABC drug exporter from an efflux transporter into a drug uptake pump would constitute a paradigm shift in efforts to overcome cancer drug resistance.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transporte Biológico/fisiología , Resistencia a Múltiples Medicamentos/fisiología , Preparaciones Farmacéuticas/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Sustitución de Aminoácidos/fisiología , Animales , Sitios de Unión/fisiología , Línea Celular , Línea Celular Tumoral , Resistencia a Antineoplásicos/fisiología , Células HeLa , Humanos , Insectos , Simulación del Acoplamiento Molecular/métodos , Rodamina 123/metabolismo , Especificidad por Sustrato/fisiología
7.
Sci Rep ; 10(1): 20614, 2020 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-33244017

RESUMEN

Sphingobium sp. strain SYK-6 is an alphaproteobacterial degrader of lignin-derived aromatic compounds, which can degrade all the stereoisomers of ß-aryl ether-type compounds. SYK-6 cells convert four stereoisomers of guaiacylglycerol-ß-guaiacyl ether (GGE) into two enantiomers of α-(2-methoxyphenoxy)-ß-hydroxypropiovanillone (MPHPV) through GGE α-carbon atom oxidation by stereoselective Cα-dehydrogenases encoded by ligD, ligL, and ligN. The ether linkages of the resulting MPHPV enantiomers are cleaved by stereoselective glutathione (GSH) S-transferases (GSTs) encoded by ligF, ligE, and ligP, generating (ßR/ßS)-α-glutathionyl-ß-hydroxypropiovanillone (GS-HPV) and guaiacol. To date, it has been shown that the gene products of ligG and SLG_04120 (ligQ), both encoding GST, catalyze GSH removal from (ßR/ßS)-GS-HPV, forming achiral ß-hydroxypropiovanillone. In this study, we verified the enzyme properties of LigG and LigQ and elucidated their roles in ß-aryl ether catabolism. Purified LigG showed an approximately 300-fold higher specific activity for (ßR)-GS-HPV than that for (ßS)-GS-HPV, whereas purified LigQ showed an approximately six-fold higher specific activity for (ßS)-GS-HPV than that for (ßR)-GS-HPV. Analyses of mutants of ligG, ligQ, and both genes revealed that SYK-6 converted (ßR)-GS-HPV using both LigG and LigQ, whereas only LigQ was involved in converting (ßS)-GS-HPV. Furthermore, the disruption of both ligG and ligQ was observed to lead to the loss of the capability of SYK-6 to convert MPHPV. This suggests that GSH removal from GS-HPV catalyzed by LigG and LigQ, is essential for cellular GSH recycling during ß-aryl ether catabolism.


Asunto(s)
Éter/metabolismo , Glutatión Transferasa/metabolismo , Sphingomonadaceae/metabolismo , Proteínas Bacterianas/metabolismo , Catálisis , Glutatión/metabolismo , Lignina/metabolismo , Oxidación-Reducción , Estereoisomerismo , Especificidad por Sustrato/fisiología
8.
J Struct Biol ; 212(2): 107628, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32971290

RESUMEN

Sialic acid residues found as terminal monosaccharides in various types of glycan chains in cell surface glycoproteins and glycolipids have been identified as important contributors of cell-cell interactions in normal vs. abnormal cellular behavior and are pivotal in diseases such as cancers. In vertebrates, sialic acids are attached to glycan chains by a conserved subset of sialyltransferases with different enzymatic and substrate specificities. ST6Gal I is a sialyltransferase using activated CMP-sialic acids as donor substrates to catalyze the formation of a α2,6-glycosidic bond between the sialic acid residue and the acceptor disaccharide LacNAc. Understanding sialyltransferases at the molecular and structural level shed light into their function. We present here two human ST6Gal I structures, which show for the first time the enzyme in the unliganded state and with the full donor substrate CMP-Neu5Ac bound. Comparison of these structures reveal flexibility of the catalytic loop, since in the unliganded structure Tyr354 adopts a conformation seen also as an alternate conformation in the substrate bound structure. CMP-Neu5Ac is bound with the side chain at C5 of the sugar residue directed outwards at the surface of the protein. Furthermore, the exact binding mode of the sialic acid moiety of the substrate directly involves sialylmotifs L, S and III and positions the sialylmotif VS in the immediate vicinity. We also present a model for the ternary complex of ST6Gal I with both the donor and the acceptor substrates.


Asunto(s)
Antígenos CD/química , Citidina Monofosfato/análogos & derivados , Citidina Monofosfato/química , Ácidos Siálicos/química , Sialiltransferasas/química , Animales , Humanos , Monosacáridos/química , Polisacáridos/química , Especificidad por Sustrato/fisiología , beta-D-Galactósido alfa 2-6-Sialiltransferasa
9.
J Struct Biol ; 212(1): 107576, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32682077

RESUMEN

Metabolite damage control is a critical but poorly defined aspect of cellular biochemistry, which likely involves many of the so far functionally uncharacterized protein domain (domains of unknown function; DUFs). We have determined the crystal structure of the human DUF89 protein product of the C6ORF211 gene to 1.85 Å. The crystal structure shows that the protein contains a core α-ß-α fold with an active site-bound metal ion and α-helical bundle N-terminal cap, which are both conserved features of subfamily III DUF89 domains. The biochemical activities of the human protein are conserved with those of a previously characterized budding yeast homolog, where an in vitro phosphatase activity is supported by divalent cations that include Co2+, Ni2+, Mn2+ or Mg2+. Full steady-state kinetics parameters of human DUF89 using a standard PNPP phosphatase assay revealed a six times higher catalytic efficiency in presence of Co2+ compared to Mg2+. The human enzyme targets a number of phosphate substrates similar to the budding yeast homolog, while it lacks a previously indicated methyltransferase activity. The highest activity on substrate was observed with fructose-1-phosphate, a potent glycating agent, and thus human DUF89 phosphatase activity may also play a role in limiting the buildup of phospho-glycan species and their related damaged metabolites.


Asunto(s)
Monoéster Fosfórico Hidrolasas/metabolismo , Proteína O-Metiltransferasa/metabolismo , Especificidad por Sustrato/fisiología , Sitios de Unión/fisiología , Catálisis , Humanos , Cinética , Metales/metabolismo , Polisacáridos/metabolismo , Conformación Proteica , Saccharomyces cerevisiae/metabolismo
10.
J Biol Chem ; 295(27): 9012-9020, 2020 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-32385112

RESUMEN

Eukaryotic DNA polymerase ß (Pol ß) plays an important role in cellular DNA repair, as it fills short gaps in dsDNA that result from removal of damaged bases. Since defects in DNA repair may lead to cancer and genetic instabilities, Pol ß has been extensively studied, especially its mechanisms for substrate binding and a fidelity-related conformational change referred to as "fingers closing." Here, we applied single-molecule FRET to measure distance changes associated with DNA binding and prechemistry fingers movement of human Pol ß. First, using a doubly labeled DNA construct, we show that Pol ß bends the gapped DNA substrate less than indicated by previously reported crystal structures. Second, using acceptor-labeled Pol ß and donor-labeled DNA, we visualized dynamic fingers closing in single Pol ß-DNA complexes upon addition of complementary nucleotides and derived rates of conformational changes. We further found that, while incorrect nucleotides are quickly rejected, they nonetheless stabilize the polymerase-DNA complex, suggesting that Pol ß, when bound to a lesion, has a strong commitment to nucleotide incorporation and thus repair. In summary, the observation and quantification of fingers movement in human Pol ß reported here provide new insights into the delicate mechanisms of prechemistry nucleotide selection.


Asunto(s)
ADN Polimerasa beta/metabolismo , ADN/metabolismo , Cristalografía por Rayos X/métodos , ADN Polimerasa I/química , ADN Polimerasa beta/fisiología , Reparación del ADN , Replicación del ADN , Proteínas de Unión al ADN/metabolismo , Transferencia Resonante de Energía de Fluorescencia/métodos , Humanos , Cinética , Modelos Moleculares , Conformación de Ácido Nucleico , Nucleótidos/metabolismo , Conformación Proteica , Especificidad por Sustrato/fisiología
11.
Sci Rep ; 10(1): 1680, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-32015448

RESUMEN

The investigation of substrate spectrum towards five racemic (rac-) aryl glycidyl ethers (1a-5a) indicated that E. coli/pveh3, an E. coli BL21(DE3) transformant harboring a PvEH3-encoding gene pveh3, showed the highest EH activity and enantiomeric ratio (E) towards rac-3a. For efficiently catalyzing the kinetic resolution of rac-3a, the activity and E value of PvEH3 were further improved by site-directed mutagenesis of selected residues. Based on the semi-rational design of an NC-loop in PvEH3, four single-site variants of pveh3 were amplified by PCR, and intracellularly expressed in E. coli BL21(DE3), respectively. E. coli/pveh3E134K and /pveh3T137P had the enhanced EH activities of 15.3 ± 0.4 and 16.1 ± 0.5 U/g wet cell as well as E values of 21.7 ± 1.0 and 21.2 ± 1.1 towards rac-3a. Subsequently, E. coli/pveh3E134K/T137P harboring a double-site variant gene was also constructed, having the highest EH activity of 22.4 ± 0.6 U/g wet cell and E value of 24.1 ± 1.2. The specific activity of the purified PvEH3E134K/T137P (14.5 ± 0.5 U/mg protein) towards rac-3a and its catalytic efficiency (kcat/Km of 5.67 mM-1 s-1) for (S)-3a were 1.7- and 3.54-fold those (8.4 ± 0.3 U/mg and 1.60 mM-1 s-1) of PvEH3. The gram-scale kinetic resolution of rac-3a using whole wet cells of E. coli/pveh3E134K/T137P was performed at 20 °C for 7.0 h, producing (R)-3a with 99.4% ees and 38.5 ± 1.2% yield. Additionally, the mechanism of PvEH3E134K/T137P with remarkably improved E value was analyzed by molecular docking simulation.


Asunto(s)
Cresoles/metabolismo , Epóxido Hidrolasas/metabolismo , Compuestos Epoxi/metabolismo , Phaseolus/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Catálisis , Epóxido Hidrolasas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Cinética , Simulación del Acoplamiento Molecular/métodos , Mutagénesis Sitio-Dirigida/métodos , Phaseolus/genética , Estereoisomerismo , Especificidad por Sustrato/fisiología
12.
Biochem Pharmacol ; 174: 113850, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32044355

RESUMEN

The human cytochrome P450 enzyme CYP4Z1 remains an understudied enzyme despite its association with poor prognosis and overexpression in breast cancer. Hence, CYP4Z1 has previously been suggested as an anti-breast cancer target. In the present study we employed extended mutation analysis to increase our understanding of the substrate binding mode of this enzyme. In a combined in vitro and in silico approach we show for the first time that residue Arg487 plays an important role in substrate recognition and binding of CYP4Z1. Using a large array of recombinant CYP4Z1 mutants we show that, apart from Asn381, all other postulated binding residues only play an auxiliary role in substrate recognition and binding. Different substrate interaction motifs were identified via dynamic pharmacophores (dynophores) and their impact on catalytically competent substrate binding was classified. These new insights on the substrate recognition and binding mode represent an important step towards the rational design of CYP4Z1 prodrugs and guide further investigations into the so far poorly understood physiological role of CYP4Z1.


Asunto(s)
Arginina/metabolismo , Asparagina/metabolismo , Simulación por Computador , Familia 4 del Citocromo P450/metabolismo , Arginina/química , Arginina/genética , Asparagina/química , Asparagina/genética , Sitios de Unión/fisiología , Familia 4 del Citocromo P450/química , Familia 4 del Citocromo P450/genética , Humanos , Mutación Missense/fisiología , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Especificidad por Sustrato/fisiología
13.
AAPS J ; 21(6): 107, 2019 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-31637538

RESUMEN

The multikinase inhibitor sorafenib (SOR) is used to treat patients with hepatocellular and renal carcinomas. SOR undergoes CYP-mediated biotransformation to a pharmacologically active N-oxide metabolite (SNO) that has been shown to accumulate to varying extents in individuals. Kinase inhibitors like SOR are frequently coadministered with a range of other drugs to improve the efficacy of anticancer drug therapy and to treat comorbidities. Recent evidence has suggested that SNO is more effective than SOR as an inhibitor of CYP3A4-mediated midazolam 1'-hydroxylation. CYP2D6 is also reportedly inhibited by SOR. The present study assessed the possibility that SNO might contribute to CYP2D6 inhibition. The inhibition kinetics of CYP2D6-mediated dextromethorphan O-demethylation were analyzed in human hepatic microsomes, with SNO found to be ~ 19-fold more active than SOR (Kis 1.8 ± 0.3 µM and 34 ± 11 µM, respectively). Molecular docking studies of SOR and SNO were undertaken using multiple crystal structures of CYP2D6. Both molecules mediated interactions with key amino acid residues in putative substrate recognition sites of CYP2D6. However, a larger number of H-bonding interactions was noted between the N-oxide moiety of SNO and active site residues that account for its greater inhibition potency. These findings suggest that SNO has the potential to contribute to pharmacokinetic interactions involving SOR, perhaps in those individuals in whom SNO accumulates.


Asunto(s)
Antineoplásicos/metabolismo , Inhibidores del Citocromo P-450 CYP2D6/metabolismo , Citocromo P-450 CYP2D6/metabolismo , Microsomas Hepáticos/metabolismo , Óxidos/metabolismo , Sorafenib/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacología , Citocromo P-450 CYP2D6/química , Inhibidores del Citocromo P-450 CYP2D6/química , Inhibidores del Citocromo P-450 CYP2D6/farmacología , Humanos , Microsomas Hepáticos/efectos de los fármacos , Óxidos/química , Óxidos/farmacología , Sorafenib/química , Sorafenib/farmacología , Especificidad por Sustrato/efectos de los fármacos , Especificidad por Sustrato/fisiología
14.
Mol Pharmacol ; 96(5): 629-640, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31515284

RESUMEN

The ATP-binding cassette transporter P-glycoprotein (P-gp) is known to limit both brain penetration and oral bioavailability of many chemotherapy drugs. Although US Food and Drug Administration guidelines require that potential interactions of investigational drugs with P-gp be explored, often this information does not enter the literature. In response, we developed a high-throughput screen to identify substrates of P-gp from a series of chemical libraries, testing a total of 10,804 compounds, most of which have known mechanisms of action. We used the CellTiter-Glo viability assay to test library compounds against parental KB-3-1 human cervical adenocarcinoma cells and the colchicine-selected subline KB-8-5-11 that overexpresses P-gp. KB-8-5-11 cells were also tested in the presence of a P-gp inhibitor (tariquidar) to assess reversibility of transporter-mediated resistance. Of the tested compounds, a total of 90 P-gp substrates were identified, including 55 newly identified compounds. Substrates were confirmed using an orthogonal killing assay against human embryonic kidney-293 cells overexpressing P-gp. We confirmed that AT7159 (cyclin-dependent kinase inhibitor), AT9283, (Janus kinase 2/3 inhibitor), ispinesib (kinesin spindle protein inhibitor), gedatolisib (PKI-587, phosphoinositide 3-kinase/mammalian target of rampamycin inhibitor), GSK-690693 (AKT inhibitor), and KW-2478 (heat-shock protein 90 inhibitor) were substrates. In addition, we assessed direct ATPase stimulation. ABCG2 was also found to confer high levels of resistance to AT9283, GSK-690693, and gedatolisib, whereas ispinesib, AT7519, and KW-2478 were weaker substrates. Combinations of P-gp substrates and inhibitors were assessed to demonstrate on-target synergistic cell killing. These data identified compounds whose oral bioavailability or brain penetration may be affected by P-gp. SIGNIFICANCE STATEMENT: The ATP-binding cassette transporter P-glycoprotein (P-gp) is known to be expressed at barrier sites, where it acts to limit oral bioavailability and brain penetration of substrates. In order to identify novel compounds that are transported by P-gp, we developed a high-throughput screen using the KB-3-1 cancer cell line and its colchicine-selected subline KB-8-5-11. We screened the Mechanism Interrogation Plate (MIPE) library, the National Center for Advancing Translational Science (NCATS) pharmaceutical collection (NPC), the NCATS Pharmacologically Active Chemical Toolbox (NPACT), and a kinase inhibitor library comprising 977 compounds, for a total of 10,804 compounds. Of the 10,804 compounds screened, a total of 90 substrates were identified of which 55 were novel. P-gp expression may adversely affect the oral bioavailability or brain penetration of these compounds.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Citotoxinas/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Proteínas de Neoplasias/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/química , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Citotoxinas/química , Citotoxinas/farmacología , Relación Dosis-Respuesta a Droga , Células HEK293 , Células HeLa , Humanos , Especificidad por Sustrato/efectos de los fármacos , Especificidad por Sustrato/fisiología
15.
Proc Natl Acad Sci U S A ; 116(39): 19513-19522, 2019 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-31492816

RESUMEN

TRiC/CCT assists the folding of ∼10% of cytosolic proteins through an ATP-driven conformational cycle and is essential in maintaining protein homeostasis. Here, we determined an ensemble of cryo-electron microscopy (cryo-EM) structures of yeast TRiC at various nucleotide concentrations, with 4 open-state maps resolved at near-atomic resolutions, and a closed-state map at atomic resolution, revealing an extra layer of an unforeseen N-terminal allosteric network. We found that, during TRiC ring closure, the CCT7 subunit moves first, responding to nucleotide binding; CCT4 is the last to bind ATP, serving as an ATP sensor; and CCT8 remains ADP-bound and is hardly involved in the ATPase-cycle in our experimental conditions; overall, yeast TRiC consumes nucleotide in a 2-ring positively coordinated manner. Our results depict a thorough picture of the TRiC conformational landscape and its allosteric transitions from the open to closed states in more structural detail and offer insights into TRiC subunit specificity in ATP consumption and ring closure, and potentially in substrate processing.


Asunto(s)
Chaperonina con TCP-1/metabolismo , Chaperonina con TCP-1/ultraestructura , Adenosina Trifosfatasas/metabolismo , Chaperonina con TCP-1/fisiología , Chaperoninas/metabolismo , Microscopía por Crioelectrón/métodos , Modelos Moleculares , Conformación Molecular , Pliegue de Proteína , Subunidades de Proteína/metabolismo , Proteostasis , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Relación Estructura-Actividad , Especificidad por Sustrato/fisiología
16.
PLoS Comput Biol ; 15(9): e1006909, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31479443

RESUMEN

Proteases are multifunctional, promiscuous enzymes that degrade proteins as well as peptides and drive important processes in health and disease. Current technology has enabled the construction of libraries of peptide substrates that detect protease activity, which provides valuable biological information. An ideal library would be orthogonal, such that each protease only hydrolyzes one unique substrate, however this is impractical due to off-target promiscuity (i.e., one protease targets multiple different substrates). Therefore, when a library of probes is exposed to a cocktail of proteases, each protease activates multiple probes, producing a convoluted signature. Computational methods for parsing these signatures to estimate individual protease activities primarily use an extensive collection of all possible protease-substrate combinations, which require impractical amounts of training data when expanding to search for more candidate substrates. Here we provide a computational method for estimating protease activities efficiently by reducing the number of substrates and clustering proteases with similar cleavage activities into families. We envision that this method will be used to extract meaningful diagnostic information from biological samples.


Asunto(s)
Biología Computacional/métodos , Péptido Hidrolasas , Análisis de Secuencia de Proteína/métodos , Especificidad por Sustrato/fisiología , Análisis por Conglomerados , Humanos , Cinética , Modelos Moleculares , Péptido Hidrolasas/análisis , Péptido Hidrolasas/química , Péptido Hidrolasas/clasificación , Péptido Hidrolasas/metabolismo , Péptidos/análisis , Péptidos/química , Péptidos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
17.
J Pharmacol Exp Ther ; 371(2): 320-326, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31455631

RESUMEN

CC-115, a triazole-containing compound, is a dual mammalian target of rapamycin (mTOR)/DNA-dependent protein kinase (DNA-PK) inhibitor currently in clinical trials. To develop this compound further, we investigated factors that may affect cellular response to CC-115. Previously, fatty acid synthase (FASN) was shown to upregulate DNA-PK activity and contribute to drug resistance; therefore, we hypothesized that FASN may affect cellular response to CC-115. Instead, however, we showed that CC-115 is a substrate of ATP-binding cassette G2 (ABCG2), a member of the ATP-binding cassette transporter superfamily, and that expression of ABCG2, not FASN, affects the potency of CC-115. ABCG2 overexpression significantly increases resistance to CC-115. Inhibiting ABCG2 function, using small-molecule inhibitors, sensitizes cancer cells to CC-115. We also found that CC-115 may be a substrate of ABCB1, another known ABC protein that contributes to drug resistance. These findings suggest that expression of ABC transporters, including ABCB1 and ABCG2, may affect the outcome in clinical trials testing CC-115. Additionally, the data indicate that ABC transporters may be used as markers for future precision use of CC-115. SIGNIFICANCE STATEMENT: In this article, we report our findings on the potential mechanism of resistance to CC-115, a dual inhibitor of mTOR and DNA-PK currently in clinical trials. We show that CC-115 is a substrate of ABCG2 and can be recognized by ABCB1, which contributes to CC-115 resistance. These findings provide novel information and potential guidance on future clinical testing of CC-115.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Resistencia a Medicamentos/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Pirazinas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Triazoles/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Ensayos Clínicos como Asunto/métodos , ADN/antagonistas & inhibidores , ADN/metabolismo , Relación Dosis-Respuesta a Droga , Resistencia a Medicamentos/fisiología , Células HEK293 , Humanos , Células MCF-7 , Factores de Riesgo , Especificidad por Sustrato/efectos de los fármacos , Especificidad por Sustrato/fisiología
18.
J Pharm Sci ; 108(10): 3416-3424, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31254495

RESUMEN

PHT2, a member of the proton-coupled oligopeptide transporter family, participates in the transportation of small peptides and histidine from lysosomes to the cytosol. It facilitates maintenance of intracellular peptide homeostasis. However, it remains a challenge to elucidate the functional properties of PHT2 due to its localization in the lysosomal membrane. The aim of this study was to explore the transport function and substrate properties of human PHT2 (hPHT2) by transfecting Madin-Darby canine kidney cells with hPHT2 mutants to obtain stably expressed protein in the cell membrane. Using this cell model, we found that the transport activity of hPHT2 reached a maximum capacity when the extracellular pH was 5.5. hPHT2 showed relatively low affinity for Gly-Sar and relatively high affinity for d3-L-histidine, with Km values of 428 ± 88 µM and 66.9 ± 5.7 µM, respectively. Several typical substrates or inhibitors of PEPT1 and PEPT2, including valacyclovir, Gly-Gly-Gly, and cefadroxil but not 5-aminolevulinic acid or captopril, were proven to be substrates of hPHT2. However, hPHT2 showed low affinity for valacyclovir with a Km value of 5350 ± 1234 µM. In conclusion, this study established a suitable and efficient cell model to explore the function of hPHT2 in vitro and provided important information on the transport activity and substrate properties of hPHT2.


Asunto(s)
Transporte Biológico/fisiología , Histidina/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Péptidos/metabolismo , Animales , Línea Celular , Membrana Celular/metabolismo , Dipéptidos/metabolismo , Perros , Humanos , Concentración de Iones de Hidrógeno , Lisosomas/metabolismo , Células de Riñón Canino Madin Darby , Oligopéptidos/metabolismo , Protones , Especificidad por Sustrato/fisiología
19.
Toxicol Appl Pharmacol ; 378: 114642, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31254566

RESUMEN

Organic anion transporting polypeptides (OATPs) are key players of drug absorption, distribution and excretion due to their broad substrate specificity, wide tissue distribution and the involvement in drug-drug interaction. OATP1B1 is specifically localized at the basolateral membrane of human hepatocytes and serves a crucial role in the drug clearance from the body. Previous studies have shown that transmembrane domains (TMs) are essential for proper functions of OATPs. In the present study, site-directed mutagenesis was performed to study the TM1 and amino-terminus of OATP1B1. Two positively charged residues, K41 and K49, as well as a hydrophobic residue I46, in TM1 were identified to be important for the proper function of the transporter. K41A and K49A exhibited altered Km value at the high and low affinity binding sites of estrone-3- sulfate (ES), respectively; while alanine substitution of I46 showed altered Km and Vmax values for both binding components of ES. Additional replacement of K41 revealed that the positively charged property at this position is important for maintaining OATP1B1 protein level and function; while the specific side-group structure of lysine at position 49 is irreplaceable for the transporter activity. Conservative replacement of I46 with leucine also recovered the function of the transporter. In addition, studies of the amino-terminus of OATP1B1 revealed that residues ranging from 19 to 27 are essential for protein stability and substrate interaction. Therefore, the amino-terminal region, which includes TM1 and the amino-terminus of OATP1B1, is important for proper function of the membrane protein.


Asunto(s)
Transportador 1 de Anión Orgánico Específico del Hígado/metabolismo , Especificidad por Sustrato/fisiología , Secuencia de Aminoácidos , Sitios de Unión/fisiología , Transporte Biológico/fisiología , Línea Celular , Membrana Celular/metabolismo , Estrona/análogos & derivados , Estrona/metabolismo , Células HEK293 , Humanos , Cinética , Mutagénesis Sitio-Dirigida/métodos , Péptidos/metabolismo , Dominios Proteicos/fisiología
20.
J Biol Chem ; 294(28): 10928-10941, 2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-31147443

RESUMEN

Cytochrome P450 (P450) enzymes are major catalysts involved in the oxidations of most drugs, steroids, carcinogens, fat-soluble vitamins, and natural products. The binding of substrates to some of the 57 human P450s and other mammalian P450s is more complex than a two-state system and has been proposed to involve mechanisms such as multiple ligand occupancy, induced-fit, and conformational-selection. Here, we used kinetic analysis of binding with multiple concentrations of substrates and computational modeling of these data to discern possible binding modes of several human P450s. We observed that P450 2D6 binds its ligand rolapitant in a mechanism involving conformational-selection. P450 4A11 bound the substrate lauric acid via conformational-selection, as did P450 2C8 with palmitic acid. Binding of the steroid progesterone to P450 21A2 was also best described by a conformational-selection model. Hexyl isonicotinate binding to P450 2E1 could be described by either a conformational-selection or an induced-fit model. Simulation of the binding of the ligands midazolam, bromocriptine, testosterone, and ketoconazole to P450 3A4 was consistent with an induced-fit or a conformational-selection model, but the concentration dependence of binding rates for varying both P450 3A4 and midazolam concentrations revealed discordance in the parameters, indicative of conformational-selection. Binding of the P450s 2C8, 2D6, 3A4, 4A11, and 21A2 was best described by conformational-selection, and P450 2E1 appeared to fit either mode. These findings highlight the complexity of human P450-substrate interactions and that conformational-selection is a dominant feature of many of these interactions.


Asunto(s)
Citocromo P-450 CYP3A/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Conformación Proteica/efectos de los fármacos , Catálisis , Citocromo P-450 CYP2D6/metabolismo , Citocromo P-450 CYP2E1/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Sistema Enzimático del Citocromo P-450/fisiología , Humanos , Cinética , Ácidos Láuricos , Ligandos , Conformación Molecular , Oxidación-Reducción , Ácido Palmítico , Unión Proteica/fisiología , Compuestos de Espiro , Especificidad por Sustrato/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA