Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Pharmacol ; 100(6): 599-608, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34599072

RESUMEN

Organic solute transporter α/ß (OSTα/ß) is a bidirectional bile acid transporter localized on the basolateral membrane of hepatic, intestinal, and renal epithelial cells. OSTα/ß plays a critical role in intestinal bile acid reabsorption and is upregulated in hepatic diseases characterized by elevated bile acids, whereas genetic variants in SLC51A/B have been associated with clinical cholestasis. OSTα/ß also transports and is inhibited by commonly used medications. However, there is currently no high-resolution structure of OSTα/ß, and structure-function data for OSTα, the proposed substrate-binding subunit, are lacking. The present study addressed this knowledge gap and identified amino acids in OSTα that are important for bile acid transport. This was accomplished using computational modeling and site-directed mutagenesis of the OSTα subunit to generate OSTα/ß mutant cell lines. Out of the 10 OSTα/ß mutants investigated, four (S228K, T229S, Q269E, Q269K) exhibited decreased [3H]-taurocholate (TCA) uptake (ratio of geometric means relative to OSTα/ß wild type (WT) of 0.76, 0.75, 0.79, and 0.13, respectively). Three OSTα/ß mutants (S228K, Q269K, E305A) had reduced [3H]-TCA efflux % (ratio of geometric means relative to OSTα/ß WT of 0.86, 0.65, and 0.79, respectively). Additionally, several OSTα/ß mutants demonstrated altered expression and cellular localization when compared with OSTα/ß WT. In summary, we identified OSTα residues (Ser228, Thr229, Gln269, Glu305) in predicted transmembrane domains that affect expression of OSTα/ß and may influence OSTα/ß-mediated bile acid transport. These data advance our understanding of OSTα/ß structure/function and can inform future studies designed to gain further insight into OSTα/ß structure or to identify additional OSTα/ß substrates and inhibitors. SIGNIFICANCE STATEMENT: OSTα/ß is a clinically important transporter involved in enterohepatic bile acid recycling with currently no high-resolution protein structure and limited structure-function data. This study identified four OSTα amino acids (Ser228, Thr229, Gln269, Glu305) that affect expression of OSTα/ß and may influence OSTα/ß-mediated bile acid transport. These data can be utilized to inform future investigation of OSTα/ß structure and refine molecular modeling approaches to facilitate the identification of substrates and/or inhibitors of OSTα/ß.


Asunto(s)
Proteínas Portadoras/química , Glicoproteínas de Membrana/química , Proteínas de Transporte de Membrana/química , Sustitución de Aminoácidos , Sitios de Unión , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células HEK293 , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Simulación de Dinámica Molecular , Unión Proteica , Ácido Taurocólico/química , Ácido Taurocólico/metabolismo
2.
J Lipid Res ; 61(11): 1524-1535, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32718973

RESUMEN

Bile acids (BAs) have been established as ubiquitous regulatory molecules implicated in a large variety of healthy and pathological processes. However, the scope of BA heterogeneity is often underrepresented in current literature. This is due in part to inadequate detection methods, which fail to distinguish the individual constituents of the BA pool. Thus, the primary aim of this study was to develop a method that would allow the simultaneous analysis of specific C24 BA species, and to apply that method to biological systems of interest. Herein, we describe the generation and validation of an LC-MS/MS assay for quantification of numerous BAs in a variety of cell systems and relevant biofluids and tissue. These studies included the first baseline level assessment for planar BAs, including allocholic acid, in cell lines, biofluids, and tissue in a nonhuman primate (NHP) laboratory animal, Macaca mulatta, in healthy conditions. These results indicate that immortalized cell lines make poor models for the study of BA synthesis and metabolism, whereas human primary hepatocytes represent a promising alternative model system. We also characterized the BA pool of M. mulatta in detail. Our results support the use of NHP models for the study of BA metabolism and pathology in lieu of murine models. Moreover, the method developed here can be applied to the study of common and planar C24 BA species in other systems.


Asunto(s)
Ácidos y Sales Biliares/análisis , Bilis/química , Hepatocitos/química , Animales , Ácidos y Sales Biliares/metabolismo , Células Cultivadas , Cromatografía Líquida de Alta Presión , Humanos , Macaca mulatta , Espectrometría de Masas en Tándem
3.
Mol Pharmacol ; 96(3): 345-354, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31436536

RESUMEN

Phenobarbital (PB), a broadly used antiseizure drug, was the first to be characterized as an inducer of cytochrome P450 by activation of the constitutive androstane receptor (CAR). Although PB is recognized as a conserved CAR activator among species via a well-documented indirect activation mechanism, conflicting results have been reported regarding PB regulation of the pregnane X receptor (PXR), a sister receptor of CAR, and the underlying mechanisms remain elusive. Here, we show that in a human CAR (hCAR)-knockout (KO) HepaRG cell line, PB significantly induces the expression of CYP2B6 and CYP3A4, two shared target genes of hCAR and human PXR (hPXR). In human primary hepatocytes and hCAR-KO HepaRG cells, PB-induced expression of CYP3A4 was markedly repressed by genetic knockdown or pharmacological inhibition of hPXR. Mechanistically, PB concentration dependently activates hPXR but not its mouse counterpart in cell-based luciferase assays. Mammalian two-hybrid assays demonstrated that PB selectively increases the functional interaction between the steroid receptor coactivator-1 and hPXR but not mouse PXR. Moreover, surface plasmon resonance binding affinity assay showed that PB directly binds to the ligand binding domain of hPXR (KD = 1.42 × 10-05). Structure-activity analysis further revealed that the amino acid tryptophan-299 within the ligand binding pocket of hPXR plays a key role in the agonistic binding of PB and mutation of tryptophan-299 disrupts PB activation of hPXR. Collectively, these data reveal that PB, a selective mouse CAR activator, activates both hCAR and hPXR, and provide novel mechanistic insights for PB-mediated activation of hPXR.


Asunto(s)
Fenobarbital/farmacología , Receptor X de Pregnano/química , Receptor X de Pregnano/genética , Receptores Citoplasmáticos y Nucleares/genética , Animales , Células Cultivadas , Receptor de Androstano Constitutivo , Citocromo P-450 CYP2B6/metabolismo , Citocromo P-450 CYP3A/metabolismo , Técnicas de Inactivación de Genes , Humanos , Ratones , Receptor X de Pregnano/metabolismo , Unión Proteica , Receptores Citoplasmáticos y Nucleares/metabolismo , Especificidad de la Especie , Resonancia por Plasmón de Superficie , Triptófano/metabolismo
4.
Mol Pharm ; 16(8): 3569-3576, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31194565

RESUMEN

The human apical sodium-dependent bile acid transporter (hASBT; SLC10A2) is responsible for the reclamation of bile acids from the intestinal lumen, providing a primary mechanism for bile acid and cholesterol homeostasis. However, the regulation of hASBT at the post-translational level is not well understood. In the present study, we investigated the role of Src family kinases (SFKs) and protein tyrosine phosphatases (PTPs) in the regulation of surface expression and function of hASBT. Inhibition of Src family kinases, via treatment with PP2, significantly reduced hASBT function, while the inhibition of PTPs by activated orthovanadate significantly induced function. Src family kinase inhibition by PP2 was associated with a concomitant decrease in maximum transport velocity (Jmax) correlated with a decrease in hASBT surface expression. Interestingly, PP2-mediated suppression of hASBT protein expression was rescued by the proteasome inhibitor MG132, suggesting that dephosphorylation impacts protein stability with the subsequent proteasome-dependent degradation of hASBT. Consequently, single-point mutations were introduced at five intracellular tyrosine residues: Y148F, Y216F, Y308F, Y311F, and Y337F. Although all mutants had significantly altered hASBT function without changes in total cellular expression, sequential tyrosine mutations at the five residues above rendered hASBT nonfunctional with diminished protein expression. Furthermore, orthovanadate-induced transport activity of single-point tyrosine mutants suggested a role for multiple tyrosine residues in the regulation of hASBT function and membrane expression. Overall, our data confirms that tyrosine phosphorylation mediated by Src family kinases (SFKs), in particular, regulates surface expression, function, and stability of hASBT.


Asunto(s)
Membrana Celular/metabolismo , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Simportadores/metabolismo , Tirosina/metabolismo , Animales , Células COS , Células CACO-2 , Chlorocebus aethiops , Humanos , Mucosa Intestinal/metabolismo , Leupeptinas/farmacología , Mutagénesis Sitio-Dirigida , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Mutación Puntual , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica/efectos de los fármacos , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/metabolismo , Proteolisis/efectos de los fármacos , Pirimidinas/farmacología , Simportadores/genética , Tirosina/genética , Vanadatos/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
5.
Biochim Biophys Acta Biomembr ; 1860(3): 645-653, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29198943

RESUMEN

The human apical sodium-dependent bile acid transporter, hASBT/SLC10A2, plays a central role in cholesterol homeostasis via the efficient reabsorption of bile acids from the distal ileum. hASBT has been shown to self-associate in higher order complexes, but while the functional role of endogenous cysteines has been reported, their implication in the oligomerization of hASBT remains unresolved. Here, we determined the self-association architecture of hASBT by site-directed mutagenesis combined with biochemical, immunological and functional approaches. We generated a cysteine-less form of hASBT by creating point mutations at all 13 endogenous cysteines in a stepwise manner. Although Cysless hASBT had significantly reduced function correlated with lowered surface expression, it featured an extra glycosylation site that facilitated its differentiation from wt-hASBT on immunoblots. Decreased protein expression was associated with instability and subsequent proteasome-dependent degradation of Cysless hASBT protein. Chemical cross-linking of wild-type and Cysless species revealed that hASBT exists as an active dimer and/or higher order oligomer with apparently no requirement for endogenous cysteine residues. This was further corroborated by co-immunoprecipitation of differentially tagged (HA-, Flag-) wild-type and Cysless hASBT. Finally, Cysless hASBT exhibited a dominant-negative effect when co-expressed with wild-type hASBT which validated heterodimerization/oligomerization at the functional level. Combined, our data conclusively demonstrate the functional existence of hASBT dimers and higher order oligomers irrespective of cysteine-mediated covalent bonds, thereby providing greater understanding of its topological assembly at the membrane surface.


Asunto(s)
Cisteína/química , Transportadores de Anión Orgánico Sodio-Dependiente/química , Simportadores/química , Secuencia de Aminoácidos , Animales , Transporte Biológico , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Cistina/química , Genes Dominantes , Glicosilación , Humanos , Inmunoprecipitación , Mutagénesis Sitio-Dirigida , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Fragmentos de Péptidos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Multimerización de Proteína , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Simportadores/genética , Ácido Taurocólico/metabolismo
6.
Mol Pharmacol ; 92(1): 75-87, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28442602

RESUMEN

The constitutive androstane receptor (CAR) plays an important role in xenobiotic metabolism, energy homeostasis, and cell proliferation. Antagonism of the CAR represents a key strategy for studying its function and may have potential clinical applications. However, specific human CAR (hCAR) antagonists are limited and conflicting data on the activity of these compounds have been reported. 1-(2-chlorophenyl)-N-methyl-N-(1-methylpropyl)-3-isoquinolinecarboxamide (PK11195), a typical peripheral benzodiazepine receptor ligand, has been established as a potent hCAR deactivator in immortalized cells; whether it inhibits hCAR activity under physiologically relevant conditions remains unclear. Here, we investigated the effects of PK11195 on hCAR in metabolically competent human primary hepatocytes (HPH) and HepaRG cells. We show that although PK11195 antagonizes hCAR in HepG2 cells, it induces the expression of CYP2B6 and CYP3A4, targets of hCAR and the pregnane X receptor (PXR), in HPH, HepaRG, and PXR-knockout HepaRG cells. Utilizing a HPH-HepG2 coculture model, we demonstrate that inclusion of HPH converts PK11195 from an antagonist to an agonist of hCAR, and such conversion was attenuated by potent CYP3A4 inhibitor ketoconazole. Metabolically, we show that the N-desmethyl metabolite is responsible for PK11195-mediated hCAR activation by facilitating hCAR interaction with coactivators and enhancing hCAR nuclear translocation in HPHs. Structure-activity analysis revealed that N-demethylation alters the interaction of PK11195 with the binding pocket of hCAR to favor activation. Together, these results indicate that removal of a methyl group switches PK11195 from a potent antagonist of hCAR to an agonist in HPH and highlights the importance of physiologically relevant metabolism when attempting to define the biologic action of small molecules.


Asunto(s)
Isoquinolinas/química , Isoquinolinas/metabolismo , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/metabolismo , Adulto , Anciano , Niño , Técnicas de Cocultivo , Receptor de Androstano Constitutivo , Relación Dosis-Respuesta a Droga , Femenino , Células Hep G2 , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Isoquinolinas/farmacología , Masculino , Persona de Mediana Edad , Estructura Secundaria de Proteína , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Relación Estructura-Actividad
7.
Biochim Biophys Acta Biomembr ; 1859(11): 2269-2276, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28887043

RESUMEN

Bile acids are the amphipathic primary end-products of cholesterol metabolism that aid in digestion as well as participate in signal transduction in several hepatic and enteric pathways. Despite the reputation of bile acids as signaling molecules implicated in disease states such as cancer and diabetes, there remain numerous bile acid species that are weakly characterized in either physiological or pathological conditions. This review presents one such group: the flat or planar bile acids, a set of bile acids found in humans during infancy and occurring again during certain diseases. As their name implies, these molecules are structurally distinct from the typical human bile acids, retaining the planar structure of their cholesterol predecessor instead of bending or twisting at the A ring. This review defines these species of bile acids in detail and describes their presence in infancy, gestation, and in disease. The large gaps in research regarding the flat bile acids are highlighted and all available experimental knowledge collected as far as 60years ago is summarized. Further, the potential for these molecules as endogenous biomarkers of liver disease and injury is discussed. Finally, the flat bile salts found in humans are compared to the ancestral and evolutionary older bile salts, which similarly have a flat steroidal structure, as mechanisms of flat bile acid biosynthesis are explored.


Asunto(s)
Ácidos y Sales Biliares/química , Ácidos y Sales Biliares/fisiología , Enfermedad/etiología , Animales , Ácidos y Sales Biliares/clasificación , Ácidos y Sales Biliares/metabolismo , Salud , Humanos , Relación Estructura-Actividad
8.
Drug Metab Dispos ; 43(11): 1773-80, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26285765

RESUMEN

Plasma membrane monoamine transporter (PMAT) is a major uptake-2 monoamine transporter that shares extensive substrate and inhibitor overlap with organic cation transporters 1-3 (OCT1-3). Currently, there are no PMAT-specific inhibitors available that can be used in in vitro and in vivo studies to differentiate between PMAT and OCT activities. In this study, we showed that IDT307 (4-(4-(dimethylamino)phenyl)-1-methylpyridinium iodide), a fluorescent analog of 1-methyl-4-phenylpyridinium (MPP+), is a transportable substrate for PMAT and that IDT307-based fluorescence assay can be used to rapidly identify and characterize PMAT inhibitors. Using the fluorescent substrate-based assays, we analyzed the interactions of eight human immunodeficiency virus (HIV) protease inhibitors (PIs) with human PMAT and OCT1-3 in human embryonic kidney 293 (HEK293) cells stably transfected with individual transporters. Our data revealed that PMAT and OCTs exhibit distinct sensitivity and inhibition patterns toward HIV PIs. PMAT is most sensitive to PI inhibition whereas OCT2 and OCT3 are resistant. OCT1 showed an intermediate sensitivity and a distinct inhibition profile from PMAT. Importantly, lopinavir is a potent PMAT inhibitor and exhibited >120 fold selectivity toward PMAT (IC50 = 1.4 ± 0.2 µM) over OCT1 (IC50 = 174 ± 40 µM). Lopinavir has no inhibitory effect on OCT2 or OCT3 at maximal tested concentrations. Lopinavir also exhibited no or much weaker interactions with uptake-1 monoamine transporters. Together, our results reveal that PMAT and OCTs have distinct specificity exemplified by their differential interaction with HIV PIs. Further, we demonstrate that lopinavir can be used as a selective PMAT inhibitor to differentiate PMAT-mediated monoamine and organic cation transport from those mediated by OCT1-3.


Asunto(s)
Proteínas de Transporte de Nucleósido Equilibrativas/antagonistas & inhibidores , Inhibidores de la Proteasa del VIH/farmacología , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Transportador 1 de Catión Orgánico/antagonistas & inhibidores , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Relación Dosis-Respuesta a Droga , Proteínas de Transporte de Nucleósido Equilibrativas/metabolismo , Células HEK293 , Humanos , Proteínas de Transporte de Catión Orgánico/metabolismo , Transportador 1 de Catión Orgánico/metabolismo , Transportador 2 de Cátion Orgánico
9.
Drug Metab Dispos ; 43(5): 725-34, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25735837

RESUMEN

Drug-induced liver injury (DILI) is an important cause of drug toxicity. Inhibition of multidrug resistance protein 4 (MRP4), in addition to bile salt export pump (BSEP), might be a risk factor for the development of cholestatic DILI. Recently, we demonstrated that inhibition of MRP4, in addition to BSEP, may be a risk factor for the development of cholestatic DILI. Here, we aimed to develop computational models to delineate molecular features underlying MRP4 and BSEP inhibition. Models were developed using 257 BSEP and 86 MRP4 inhibitors and noninhibitors in the training set. Models were externally validated and used to predict the affinity of compounds toward BSEP and MRP4 in the DrugBank database. Compounds with a score above the median fingerprint threshold were considered to have significant inhibitory effects on MRP4 and BSEP. Common feature pharmacophore models were developed for MRP4 and BSEP with LigandScout software using a training set of nine well characterized MRP4 inhibitors and nine potent BSEP inhibitors. Bayesian models for BSEP and MRP4 inhibition/noninhibition were developed with cross-validated receiver operator curve values greater than 0.8 for the test sets, indicating robust models with acceptable false positive and false negative prediction rates. Both MRP4 and BSEP inhibitor pharmacophore models were characterized by hydrophobic and hydrogen-bond acceptor features, albeit in distinct spatial arrangements. Similar molecular features between MRP4 and BSEP inhibitors may partially explain why various drugs have affinity for both transporters. The Bayesian (BSEP, MRP4) and pharmacophore (MRP4, BSEP) models demonstrated significant classification accuracy and predictability.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Ácidos y Sales Biliares/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP , Teorema de Bayes , Colestasis/metabolismo , Simulación por Computador , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/metabolismo , Humanos , Factores de Riesgo
10.
Mol Pharm ; 12(6): 1961-9, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25950742

RESUMEN

Internalization and intracellular trafficking of dendrimer-drug conjugates play an important role in achieving successful drug delivery. In this study, we aimed to elucidate the endocytosis mechanisms and subcellular localization of poly-l-lysine (PLL) dendrimers in Caco-2 cells. We also investigated the impact of fluorophore conjugation on cytotoxicity, uptake, and transepithelial transport. Oregon green 514 (OG) was conjugated to PLL G3 at either the dendrimer periphery or the core. Chemical inhibitors of clathrin-, caveolin-, cholesterol-, and dynamin-mediated endocytosis pathways and macropinocytosis were employed to establish internalization mechanisms, while colocalization with subcellular markers was used to determine dendrimer trafficking. Cell viability, internalization, and uptake were all influenced by the site of fluorophore conjugation. Uptake was found to be highly dependent on cholesterol- and dynamin-mediated endocytosis as well as macropinocytosis. Dendrimers were trafficked to endosomes and lysosomes, and subcellular localization was impacted by the fluorophore conjugation site. The results of this study indicate that PLL dendrimers exploit multiple pathways for cellular entry, and internalization and trafficking can be impacted by conjugation. Therefore, design of dendrimer-drug conjugates requires careful consideration to achieve successful drug delivery.


Asunto(s)
Dendrímeros/química , Dendrímeros/metabolismo , Lisina/química , Transporte Biológico/fisiología , Células CACO-2 , Endocitosis/fisiología , Humanos , Nanomedicina
11.
Pharm Res ; 37(7): 135, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32642849
12.
Biochem J ; 459(2): 301-12, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24498857

RESUMEN

The sodium/bile acid co-transporter ASBT [apical sodium-dependent bile acid transporter; SLC10A2 (solute carrier family 10 member 2)] plays a key role in the enterohepatic recycling of the bile acids and indirectly contributes to cholesterol homoeostasis. ASBT inhibitors reportedly lower plasma triglyceride levels and increase HDL (high-density lipoprotein) cholesterol levels. RSV (resveratrol), a major constituent of red wine, is known to lower LDL (low-density lipoprotein) cholesterol levels, but its mechanism of action is still unclear. In the present study, we investigated the possible involvement of ASBT in RSV-mediated cholesterol-lowering effects. We demonstrate that RSV inhibits ASBT protein expression and function via a SIRT1 (sirtuin 1)-independent mechanism. The effect was specific to ASBT since other transporters involved in cholesterol homoeostasis, NTCP (SLC10A1), OSTα (SLC51A) and ABCG1 (ATP-binding cassette G1), remained unaffected. ASBT inhibition by RSV was reversed by proteasome inhibitors (MG-132 and lactacystin) and the ubiquitin inhibitor LDN57444, suggesting involvement of the ubiquitin-proteasome pathway. Immunoprecipitation revealed high levels of ubiquitinated ASBT after RSV treatment. Phosphorylation at Ser335 and Thr339 was shown previously to play a role in proteosomal degradation of rat ASBT. However, mutation at corresponding residues in rat ASBT revealed that phosphorylation does not contribute to RSV-mediated degradation of ASBT. Combined, our data indicate that RSV promotes ASBT degradation via the ubiquitin-proteasome pathway without requiring phosphorylation. We conclude that regulation of ASBT expression by RSV may have clinical relevance with regard to the observed cholesterol-lowering effects of RSV.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Proteolisis/efectos de los fármacos , Estilbenos/farmacología , Simportadores/metabolismo , Animales , Línea Celular , Humanos , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Fosforilación , Transporte de Proteínas/efectos de los fármacos , Resveratrol , Sirtuina 1/genética , Sirtuina 1/metabolismo , Simportadores/genética
13.
J Biol Chem ; 288(45): 32394-32404, 2013 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-24045943

RESUMEN

Human apical sodium-dependent bile acid transporter (hASBT, SLC10A2) is responsible for intestinal reabsorption of bile acids and plays a key role in cholesterol homeostasis. We used a targeted and systematic approach to delineate the role of highly conserved transmembrane helix 2 on the expression and function of hASBT. Cysteine mutation significantly depressed transport activity for >60% of mutants without affecting cell surface localization of the transporter. All mutants were inaccessible toward chemical modification by membrane-impermeant MTSET reagent, strongly suggesting that transmembrane 2 (TM2) plays an indirect role in bile acid substrate translocation. Both bile acid uptake and sodium dependence of TM2 mutants revealed a distinct α-helical periodicity. Kinetic studies with conservative and non-conservative mutants of sodium sensitive residues further underscored the importance of Gln(75), Phe(76), Met(79), Gly(83), Leu(86), Phe(90), and Asp(91) in hASBT function. Computational analysis indicated that Asp(91) may coordinate with sodium during the transport cycle. Combined, our data propose that a consortium of sodium-sensitive residues along with previously reported residues (Thr(134), Leu(138), and Thr(149)) from TM3 may form the sodium binding and translocation pathway. Notably, residues Gln(75), Met(79), Thr(82), and Leu(86) from TM2 are highly conserved in TM3 of a putative remote bacterial homologue (ASBTNM), suggesting a universal mechanism for the SLC10A transporter family.


Asunto(s)
Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Sodio/metabolismo , Simportadores/metabolismo , Sustitución de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Humanos , Transporte Iónico/fisiología , Cinética , Mutación Missense , Transportadores de Anión Orgánico Sodio-Dependiente/química , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Sodio/química , Simportadores/química , Simportadores/genética
14.
Drug Metab Dispos ; 42(4): 541-5, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24598705

RESUMEN

The significance of transporters in the disposition, metabolism, and elimination of drugs is well recognized. One gap in our knowledge is a comprehensive understanding of how drug transporters change functionality (their amount and activity) in response to disease and how disease and its inevitable pathology change transporter expression. In this issue of Drug Metabolism and Disposition a series of review and primary research articles are presented to highlight the importance of transporters in toxicity and disease. Because of the central role of the liver in drug metabolism, many of the articles in this theme issue focus on transporters in the liver and how pathology or alterations in physiology affects transporter expression. The contributing authors have also considered the role of transporters in drug interactions as well as drug-induced liver injury. Noninvasive approaches to assessing transporter function in vivo are also described. Several articles highlight important issues in oncology where toxicity must be balanced against efficacy. In total, this theme issue will provide a stepping-stone to future studies that will establish a more comprehensive understanding of transporters in disease.


Asunto(s)
Proteínas Portadoras/metabolismo , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/metabolismo , Preparaciones Farmacéuticas/metabolismo , Xenobióticos/farmacocinética , Animales , Proteínas Portadoras/genética , Enfermedad , Interacciones Farmacológicas , Humanos , Neoplasias/metabolismo
15.
Drug Metab Dispos ; 42(4): 665-74, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24154606

RESUMEN

Impaired hepatic bile acid export may contribute to development of cholestatic drug-induced liver injury (DILI). The multidrug resistance-associated proteins (MRP) 3 and 4 are postulated to be compensatory hepatic basolateral bile acid efflux transporters when biliary excretion by the bile salt export pump (BSEP) is impaired. BSEP inhibition is a risk factor for cholestatic DILI. This study aimed to characterize the relationship between MRP3, MRP4, and BSEP inhibition and cholestatic potential of drugs. The inhibitory effect of 88 drugs (100 µM) on MRP3- and MRP4-mediated substrate transport was measured in membrane vesicles. Drugs selected for investigation included 50 BSEP non-inhibitors (24 non-cholestatic; 26 cholestatic) and 38 BSEP inhibitors (16 non-cholestatic; 22 cholestatic). MRP4 inhibition was associated with an increased risk of cholestatic potential among BSEP non-inhibitors. In this group, for each 1% increase in MRP4 inhibition, the odds of the drug being cholestatic increased by 3.1%. Using an inhibition cutoff of 21%, which predicted a 50% chance of cholestasis, 62% of cholestatic drugs inhibited MRP4 (P < 0.05); in contrast, only 17% of non-cholestatic drugs were MRP4 inhibitors. Among BSEP inhibitors, MRP4 inhibition did not provide additional predictive value of cholestatic potential; almost all BSEP inhibitors were also MRP4 inhibitors. Inclusion of pharmacokinetic predictor variables (e.g., maximal unbound concentration in plasma) in addition to percent MRP4 inhibition in logistic regression models did not improve cholestasis prediction. Association of cholestasis with percent MRP3 inhibition was not statistically significant, regardless of BSEP-inhibition status. Inhibition of MRP4, in addition to BSEP, may be a risk factor for the development of cholestatic DILI.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Colestasis/metabolismo , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Preparaciones Farmacéuticas , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP , Transportadoras de Casetes de Unión a ATP/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Colestasis/inducido químicamente , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/etiología , Células HEK293 , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Modelos Logísticos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Preparaciones Farmacéuticas/administración & dosificación , Preparaciones Farmacéuticas/química , Valor Predictivo de las Pruebas , Factores de Riesgo , Transfección
16.
Toxicol Appl Pharmacol ; 279(1): 33-42, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24878338

RESUMEN

OBJECTIVE: Accumulating evidence suggests that activation of mouse constitutive androstane receptor (mCAR) alleviates type 2 diabetes and obesity by inhibiting hepatic gluconeogenesis, lipogenesis, and fatty acid synthesis. However, the role of human (h) CAR in energy metabolism is largely unknown. The present study aims to investigate the effects of selective hCAR activators on hepatic energy metabolism in human primary hepatocytes (HPH). METHODS: Ligand-based structure-activity models were used for virtual screening of the Specs database (www.specs.net) followed by biological validation in cell-based luciferase assays. The effects of two novel hCAR activators (UM104 and UM145) on hepatic energy metabolism were evaluated in HPH. RESULTS: Real-time PCR and Western blotting analyses reveal that activation of hCAR by UM104 and UM145 significantly repressed the expression of glucose-6-phosphatase and phosphoenolpyruvate carboxykinase, two pivotal gluconeogenic enzymes, while exerting negligible effects on the expression of genes associated with lipogenesis and fatty acid synthesis. Functional experiments show that UM104 and UM145 markedly inhibit hepatic synthesis of glucose but not triglycerides in HPH. In contrast, activation of mCAR by 1,4-bis[2-(3,5-dichloropyridyloxy)]benzene, a selective mCAR activator, repressed the expression of genes associated with gluconeogenesis, lipogenesis, and fatty acid synthesis in mouse primary hepatocytes, which were consistent with previous observations in mouse model in vivo. CONCLUSION: Our findings uncover an important species difference between hCAR and mCAR in hepatic energy metabolism, where hCAR selectively inhibits gluconeogenesis without suppressing fatty acid synthesis. IMPLICATIONS: Such species selectivity should be considered when exploring CAR as a potential therapeutic target for metabolic disorders.


Asunto(s)
Ácidos Grasos/biosíntesis , Gluconeogénesis/efectos de los fármacos , Lipogénesis/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/agonistas , Animales , Western Blotting , Línea Celular , Simulación por Computador , Receptor de Androstano Constitutivo , Metabolismo Energético/efectos de los fármacos , Glucosa/metabolismo , Glucosa-6-Fosfatasa/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Fosfoenolpiruvato Carboxiquinasa (ATP)/metabolismo , Cultivo Primario de Células , Transporte de Proteínas/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Triglicéridos/metabolismo
17.
Pharm Res ; 31(9): 2429-38, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24648136

RESUMEN

PURPOSE: Poly(amidoamine) (PAMAM) dendrimers show great promise for utilization as oral drug delivery vehicles. These polymers are capable of traversing epithelial barriers, and have been shown to translocate by both transcellular and paracellular routes. While many proof-of-concept studies have shown that PAMAM dendrimers improve intestinal transport, little information exists on the mechanisms of paracellular transport, specifically dendrimer-induced tight junction modulation. METHODS: Using anionic G3.5 and cationic G4 PAMAM dendrimers with known absorption enhancers, we investigated tight junction modulation in Caco-2 monolayers by visualization and mannitol permeability and compared dendrimer-mediated tight junction modulation to that of established permeation enhancers. [(14)C]-Mannitol permeability in the presence and absence of phospholipase C-dependent signaling pathway inhibitors was also examined and indicated that this pathway may mediate dendrimer-induced changes in permeability. RESULTS: Differences between G3.5 and G4 in tight junction protein staining and permeability with inhibitors were evident, suggesting divergent mechanisms were responsible for tight junction modulation. These dissimilarities are further intimated by the intracellular calcium release caused by G4 but not G3.5. Based on our results, it is apparent that the underlying mechanisms of dendrimer permeability are complex, and the complexities are likely a result of the density and sign of the surface charges of PAMAM dendrimers. CONCLUSIONS: The results of this study will have implications on the future use of PAMAM dendrimers for oral drug delivery.


Asunto(s)
Dendrímeros/farmacología , Manitol/farmacocinética , Nylons/farmacología , Permeabilidad/efectos de los fármacos , Células CACO-2 , Calcio/metabolismo , Humanos , Cadenas Ligeras de Miosina/metabolismo , Uniones Estrechas/metabolismo
18.
Pharm Res ; 31(2): 401-13, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24092051

RESUMEN

PURPOSE: This study investigates the cellular uptake and trafficking of liposomes in Caco-2 cells, using vesicles with distinct average diameters ranging from 40.6 nm to 276.6 nm. Liposomes were prepared by microfluidic hydrodynamic flow focusing, producing nearly-monodisperse populations and enabling size-dependent uptake to be effectively evaluated. METHODS: Populations of PEG-conjugated liposomes of various distinct sizes were prepared in a disposable microfluidic device using a simple continuous-flow microfluidic technique. Liposome cellular uptake was investigated using flow cytometry and confocal microscopy. RESULTS: Liposome uptake by Caco-2 cells was observed to be strongly size-dependent for liposomes with mean diameters ranging from 40.6 nm to 276.6 nm. When testing these liposomes against endocytosis inhibitors, cellular uptake of the largest (97.8 nm and 162.1 nm in diameter) liposomes were predominantly subjected to clathrin-dependent uptake mechanisms, the medium-sized (72.3 nm in diameter) liposomes seemed to be influenced by all investigated pathways and the smallest liposomes (40.6 nm in diameter) primarily followed a dynamin-dependent pathway. In addition, the 40.6 nm, 72.3 nm, and 162.1 nm diameter liposomes showed slightly decreased accumulation within endosomes after 1 h compared to liposomes which were 97.8 nm in diameter. Conversely, liposome co-localization with lysosomes was consistent for liposomes ranging from 40.6 nm to 97.8 nm in diameter. CONCLUSIONS: The continuous-flow synthesis of nearly-monodisperse populations of liposomes of distinct size via a microfluidic hydrodynamic flow focusing technique enabled unique in vitro studies in which specific effects of particle size on cellular uptake were elucidated. The results of this study highlight the significant influence of liposome size on cellular uptake mechanisms and may be further exploited for increasing specificity, improving efficacy, and reducing toxicity of liposomal drug delivery systems.


Asunto(s)
Liposomas/química , Liposomas/metabolismo , Células CACO-2 , Línea Celular Tumoral , Clatrina/química , Clatrina/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Dinaminas/química , Dinaminas/metabolismo , Endocitosis/fisiología , Humanos , Microfluídica/métodos , Tamaño de la Partícula , Polietilenglicoles/química , Polietilenglicoles/metabolismo
19.
Am J Pharm Educ ; 88(2): 100641, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38185465

RESUMEN

OBJECTIVE: The objective of this review is to provide the conclusions from the American Association of Colleges of Pharmacy (AACP) Council of Deans (COD) Taskforce on Research and Scholarship. FINDINGS: The charges and the findings of the committee are: (1) Define the scholarship needs/opportunities to strengthen the outputs. The committee recommends that AACP update its definitions of research/scholarship to include discovery, integration, application/practice, and teaching/learning. A deployed survey demonstrated a high Special Interest Groups research/scholarship interest. (2) Assemble a toolkit of grant and scholarship resources to assist colleges/schools. The AACP should update the existing funding opportunity listing and combine it with additional resources. (3) Create a framework for effective research collaboration and mentorship. The AACP should identify key areas of pharmacy research and experts to serve as mentors and to meet with external stakeholders. (4) and (5) Consider the need for and purpose of a COD standing committee for research and scholarship. Explore the value of a formal research dean's subcommittee. It was recommended that AACP form a research/scholarship committee or Special Interest Groups and create the Pharmacy Scholarship, Research, and Graduate Education pre-meeting to the Interim Meeting. (6) Identify key statements/outputs of the COD that need to be prepared for publication/sharing. We recommended the key statement/outputs in the areas of discovery, integration, application/practice, and teaching and learning. SUMMARY: The taskforce reviewed the state of research and scholarship across the Academy and provided recommendations with the goal of advancing research across all areas of the pharmacy profession.


Asunto(s)
Educación de Postgrado en Farmacia , Educación en Farmacia , Investigación en Farmacia , Farmacia , Estados Unidos , Humanos , Becas , Facultades de Farmacia
20.
Biochemistry ; 52(30): 5117-24, 2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23815591

RESUMEN

The human apical sodium-dependent bile acid transporter (hASBT, SLC10A2), primarily expressed in the ileum, is involved in both the recycling of bile acids and cholesterol homeostasis. In this study, the structure-function relationship of transmembrane domain 5 (TM5) residues involved in transport is elucidated. Cysteine scanning mutagenesis of each consecutive residue on TM5 resulted in 96% of mutants having a significantly decreased transport activity, although each was expressed at the cell surface. Specifically, G197 and I208 were no longer functional, and G201 and G212 functioned at a level of <10% upon cysteine mutation. Interestingly, each of these exists along one face of the helix. Studies suggest that neither G201 nor G212 is on the substrate pathway. Conservative alanine mutations of the four residues displayed a higher activity in all but G197A, indicating its functional importance. G197 and G201 form a GxxxG motif, which has been found to be important in helix-helix interactions. According to our model, G197 and G201 face transmembrane domain 4 (TM4) residues G179 and P175, respectively. Similarly, G212 faces G237, which forms part of a GxxxG domain in transmembrane domain 6 (TM6). It is possible that these GxxxG domains and their interacting partners are responsible for maintaining the structure of the helices and their interactions with one another. I205 and I208 are both in positions to anchor the GxxxG domains and direct the change in interaction of TM5 from TM4 to TM6. Combined, the results suggest that residues along TM5 are critical for ASBT function but are not directly involved in substrate translocation.


Asunto(s)
Modelos Moleculares , Transportadores de Anión Orgánico Sodio-Dependiente/química , Simportadores/química , Alanina/química , Sustitución de Aminoácidos , Animales , Sitios de Unión , Unión Competitiva , Transporte Biológico , Células COS , Chlorocebus aethiops , Cisteína/química , Humanos , Cinética , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Transportadores de Anión Orgánico Sodio-Dependiente/antagonistas & inhibidores , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Sodio/metabolismo , Simportadores/antagonistas & inhibidores , Simportadores/genética , Simportadores/metabolismo , Ácido Taurocólico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA