Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 298(6): 101974, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35469921

RESUMO

Organic cation transporter 1 (OCT1) is a membrane transporter that affects hepatic uptake of cationic and weakly basic drugs. OCT1 transports structurally highly diverse substrates. The mechanisms conferring this polyspecificity are unknown. Here, we analyzed differences in transport kinetics between human and mouse OCT1 orthologs to identify amino acids that contribute to the polyspecificity of OCT1. Following stable transfection of HEK293 cells, we observed more than twofold differences in the transport kinetics of 22 out of 28 tested substrates. We found that the ß2-adrenergic drug fenoterol was transported with eightfold higher affinity but at ninefold lower capacity by human OCT1. In contrast, the anticholinergic drug trospium was transported with 11-fold higher affinity but at ninefold lower capacity by mouse Oct1. Using human-mouse chimeric constructs and site-directed mutagenesis, we identified nonconserved amino acids Cys36 and Phe32 as responsible for the species-specific differences in fenoterol and trospium uptake. Substitution of Cys36 (human) to Tyr36 (mouse) caused a reversal of the affinity and capacity of fenoterol but not trospium uptake. Substitution of Phe32 to Leu32 caused reversal of trospium but not fenoterol uptake kinetics. Comparison of the uptake of structurally similar ß2-adrenergics and molecular docking analyses indicated the second phenol ring, 3.3 to 4.8 Å from the protonated amino group, as essential for the affinity for fenoterol conferred by Cys36. This is the first study to report single amino acids as determinants of OCT1 polyspecificity. Our findings suggest that structure-function data of OCT1 is not directly transferrable between substrates or species.


Assuntos
Proteínas da Membrana Plasmática de Transporte de Catecolaminas/química , Transportador 1 de Cátions Orgânicos , Sequência de Aminoácidos , Animais , Proteínas da Membrana Plasmática de Transporte de Catecolaminas/metabolismo , Fenoterol , Células HEK293 , Humanos , Camundongos , Simulação de Acoplamento Molecular , Transportador 1 de Cátions Orgânicos/química , Transportador 1 de Cátions Orgânicos/metabolismo
2.
Drug Metab Dispos ; 48(12): 1380-1392, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33037045

RESUMO

The most commonly used oral antidiabetic drug, metformin, is a substrate of the hepatic uptake transporter OCT1 (gene name SLC22A1). However, OCT1 deficiency leads to more pronounced reductions of metformin concentrations in mouse than in human liver. Similarly, the effects of OCT1 deficiency on the pharmacokinetics of thiamine were reported to differ between human and mouse. Here, we compared the uptake characteristics of metformin and thiamine between human and mouse OCT1 using stably transfected human embryonic kidney 293 cells. The affinity for metformin was 4.9-fold lower in human than in mouse OCT1, resulting in a 6.5-fold lower intrinsic clearance. Therefore, the estimated liver-to-blood partition coefficient is only 3.34 in human compared with 14.4 in mouse and may contribute to higher intrahepatic concentrations in mice. Similarly, the affinity for thiamine was 9.5-fold lower in human than in mouse OCT1. Using human-mouse chimeric OCT1, we showed that simultaneous substitution of transmembrane helices TMH2 and TMH3 resulted in the reversal of affinity for metformin. Using homology modeling, we suggest several explanations, of which a different interaction of Leu155 (human TMH2) compared with Val156 (mouse TMH2) with residues in TMH3 had the strongest experimental support. In conclusion, the contribution of human OCT1 to the cellular uptake of thiamine and especially of metformin may be much lower than that of mouse OCT1. This may lead to an overestimation of the effects of OCT1 on hepatic concentrations in humans when using mouse as a model. In addition, comparative analyses of human and mouse orthologs may help reveal mechanisms of OCT1 transport. SIGNIFICANCE STATEMENT: OCT1 is a major hepatic uptake transporter of metformin and thiamine, but this study reports strong differences in the affinity for both compounds between human and mouse OCT1. Consequently, intrahepatic metformin concentrations could be much higher in mice than in humans, impacting metformin actions and representing a strong limitation of using rodent animal models for predictions of OCT1-related pharmacokinetics and efficacy in humans. Furthermore, OCT1 transmembrane helices TMH2 and TMH3 were identified to confer the observed species-specific differences in metformin affinity.


Assuntos
Metformina/farmacocinética , Transportador 1 de Cátions Orgânicos/metabolismo , Tiamina/farmacocinética , Animais , Avaliação Pré-Clínica de Medicamentos/métodos , Células HEK293 , Hepatócitos , Humanos , Fígado/enzimologia , Masculino , Camundongos , Transportador 1 de Cátions Orgânicos/genética , Transportador 1 de Cátions Orgânicos/ultraestrutura , Conformação Proteica em alfa-Hélice/genética , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/ultraestrutura , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Relação Estrutura-Atividade
3.
Clin Pharmacol Ther ; 105(1): 190-200, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29882324

RESUMO

Cycloguanil, the active metabolite of proguanil, acts on malaria schizonts in erythrocytes and hepatocytes. We analyzed the impact of the organic cation transporter OCT1 on hepatocellular uptake and pharmacokinetics of proguanil and cycloguanil. OCT1 transported both proguanil and cycloguanil. Common variants OCT1*3 and OCT1*4 caused a substantial decrease and OCT1*5 and OCT1*6 complete abolishment of proguanil uptake. In 39 healthy subjects, low-activity variants OCT1*3 and OCT1*4 had only minor effects on proguanil pharmacokinetics. However, both, cycloguanil area under the time-concentration curve and the cycloguanil-to-proguanil ratio were significantly dependent on number of these low-functional alleles (P = 0.02 for both). Together, CYP2C19, CYP3A5, OCT1 polymorphisms, and sex accounted for 61% of the variation in the cycloguanil-to-proguanil ratio. Most importantly, in vitro OCT1 inhibition caused a fivefold decrease of intracellular cycloguanil concentrations in primary human hepatocytes. In conclusion, OCT1-mediated uptake is a limiting step in bioactivation of proguanil, and OCT1 polymorphisms may affect proguanil efficacy against hepatic malaria schizonts.


Assuntos
Antimaláricos/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Fator 1 de Transcrição de Octâmero/deficiência , Proguanil/metabolismo , Triazinas/metabolismo , Adolescente , Adulto , Antimaláricos/farmacocinética , Relação Dose-Resposta a Droga , Feminino , Células HEK293 , Humanos , Masculino , Pessoa de Meia-Idade , Proguanil/farmacocinética , Triazinas/farmacocinética , Adulto Jovem
4.
Clin Pharmacol Ther ; 103(5): 868-878, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-28791698

RESUMO

Fenoterol is a widely used anti-asthmatic and tocolytic agent, but high plasma concentrations of fenoterol may lead to severe and even fatal adverse reactions. We studied whether heritable deficiency of the liver organic cation transporter 1 (OCT1), a trait observed in 3% of Europeans and white Americans, affects fenoterol plasma concentrations and toxicity. OCT1 transported fenoterol with high affinity, and OCT1 inhibition in human hepatocytes reduced fenoterol uptake threefold. After administration of 180 µg of fenoterol to 39 healthy individuals, the OCT1-deficient individuals (zero active OCT1 alleles; n = 5) showed 1.9-fold greater systemic fenoterol exposure (P = 4.0 × 10-5 ) and 1.7-fold lower volume of distribution (P = 8.0 × 10-5 ). Correspondingly, the OCT1-deficient individuals had a 1.5-fold stronger increase in heart rate (P = 0.002), a 3.4-fold greater increase in blood glucose (P = 3.0 × 10-5 ), and significantly lower serum potassium levels. In conclusion, heritable OCT1 deficiency significantly increases plasma concentrations of fenoterol and may be an important factor underlying the excess mortality associated with fenoterol.


Assuntos
Sistema Cardiovascular/efeitos dos fármacos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/etiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/metabolismo , Fenoterol/efeitos adversos , Doenças Metabólicas/induzido quimicamente , Fator 1 de Transcrição de Octâmero/deficiência , Alelos , Transporte Biológico/efeitos dos fármacos , Glicemia/efeitos dos fármacos , Sistema Cardiovascular/metabolismo , Células HEK293 , Frequência Cardíaca/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Doenças Metabólicas/metabolismo , Potássio/sangue
5.
PLoS One ; 12(12): e0189521, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29236753

RESUMO

BACKGROUND: Ranitidine (Zantac®) is a H2-receptor antagonist commonly used for the treatment of acid-related gastrointestinal diseases. Ranitidine was reported to be a substrate of the organic cation transporters OCT1 and OCT2. The hepatic transporter OCT1 is highly genetically variable. Twelve major alleles confer partial or complete loss of OCT1 activity. The effects of these polymorphisms are highly substrate-specific and therefore difficult to predict. The renal transporter OCT2 has a common polymorphism, Ala270Ser, which was reported to affect OCT2 activity. AIM: In this study we analyzed the effects of genetic polymorphisms in OCT1 and OCT2 on the uptake of ranitidine and on its potency to inhibit uptake of other drugs. METHODS AND RESULTS: We characterized ranitidine uptake using HEK293 and CHO cells stably transfected to overexpress wild type OCT1, OCT2, or their naturally occurring allelic variants. Ranitidine was transported by wild-type OCT1 with a Km of 62.9 µM and a vmax of 1125 pmol/min/mg protein. Alleles OCT1*5, *6, *12, and *13 completely lacked ranitidine uptake. Alleles OCT1*2, *3, *4, and *10 had vmax values decreased by more than 50%. In contrast, OCT1*8 showed an increase of vmax by 25%. The effects of OCT1 alleles on ranitidine uptake strongly correlated with the effects on morphine uptake suggesting common interaction mechanisms of both drugs with OCT1. Ranitidine inhibited the OCT1-mediated uptake of metformin and morphine at clinically relevant concentrations. The inhibitory potency for morphine uptake was affected by the OCT1*2 allele. OCT2 showed only a limited uptake of ranitidine that was not significantly affected by the Ala270Ser polymorphism. CONCLUSIONS: We confirmed ranitidine as an OCT1 substrate and demonstrated that common genetic polymorphisms in OCT1 strongly affect ranitidine uptake and modulate ranitidine's potential to cause drug-drug interactions. The effects of the frequent OCT1 polymorphisms on ranitidine pharmacokinetics in humans remain to be analyzed.


Assuntos
Antagonistas dos Receptores H2 da Histamina/farmacocinética , Transportador 1 de Cátions Orgânicos/metabolismo , Transportador 2 de Cátion Orgânico/metabolismo , Polimorfismo Genético , Ranitidina/farmacocinética , Animais , Células CHO , Cricetulus , Células HEK293 , Humanos , Transportador 1 de Cátions Orgânicos/genética , Transportador 2 de Cátion Orgânico/genética
7.
Biol Chem ; 398(2): 237-249, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27676604

RESUMO

Tropane alkaloids and their derivatives are anticholinergic drugs with narrow therapeutic range. Here we characterize the organic cation transporters from the SLC22 (OCT1, OCT2, and OCT3) and the SLC47 families (MATE1 and MATE2-K) as potential mediators of the renal and extra-renal excretion, the two major roads of elimination of these substances. All analyzed compounds inhibited and the quaternary amine derivatives ipratropium and trospium were strongly transported by OCTs and MATEs. Overexpression of OCTs or MATEs in HEK293 cells resulted in an up to 63-fold increase in the uptake of ipratropium (Km of 0.32 µm to OCT2 and Vmax of 3.34 nmol×mg protein-1×min-1 to MATE1). The transcellular transport of ipratropium was 16-fold higher in OCT2-MATE1 and 10-fold higher in OCT1-MATE1 overexpressing compared to control MDCKII cells. Genetic polymorphisms in OCT1 and OCT2 affected ipratropium uptake and clinically relevant concentration of ondansetron and pyrithiamine inhibited ipratropium uptake via MATEs by more than 90%. This study suggests that OCT1, OCT2 and MATEs may be strongly involved in the renal and extra-renal elimination of ipratropium and other quaternary amine alkaloids. These substances have a notoriously narrow therapeutic range and the drug-drug interactions suggested here should be further critically evaluated in humans.


Assuntos
Proteínas da Membrana Plasmática de Transporte de Catecolaminas/antagonistas & inibidores , Proteínas da Membrana Plasmática de Transporte de Catecolaminas/metabolismo , Proteínas de Transporte de Cátions Orgânicos/antagonistas & inibidores , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Tropanos/metabolismo , Tropanos/farmacologia , Animais , Transporte Biológico/efeitos dos fármacos , Proteínas da Membrana Plasmática de Transporte de Catecolaminas/genética , Permeabilidade da Membrana Celular/efeitos dos fármacos , Cães , Interações Medicamentosas , Células HEK293 , Humanos , Ipratrópio/metabolismo , Células Madin Darby de Rim Canino , Proteínas de Transporte de Cátions Orgânicos/genética , Polimorfismo de Nucleotídeo Único , Tropanos/química
8.
Genome Med ; 7(1): 56, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26157489

RESUMO

BACKGROUND: The organic cation transporter OCT1 (SLC22A1) mediates the uptake of vitamin B1, cationic drugs, and xenobiotics into hepatocytes. Nine percent of Caucasians lack or have very low OCT1 activity due to loss-of-function polymorphisms in OCT1 gene. Here we analyzed the global genetic variability in OCT1 to estimate the therapeutic relevance of OCT1 polymorphisms in populations beyond Caucasians and to identify evolutionary patterns of the common loss of OCT1 activity in humans. METHODS: We applied massively parallel sequencing to screen for coding polymorphisms in 1,079 unrelated individuals from 53 populations worldwide. The obtained data was combined with the existing 1000 Genomes data comprising an additional 1,092 individuals from 14 populations. The identified OCT1 variants were characterized in vitro regarding their cellular localization and their ability to transport 10 known OCT1 substrates. Both the population genetics data and transport data were used in tandem to generate a world map of loss of OCT1 activity. RESULTS: We identified 16 amino acid substitutions potentially causing loss of OCT1 function and analyzed them together with five amino acid substitutions that were not expected to affect OCT1 function. The variants constituted 16 major alleles and 14 sub-alleles. Six major alleles showed improper subcellular localization leading to substrate-wide loss in activity. Five major alleles showed correct subcellular localization, but substrate-specific loss of activity. Striking differences were observed in the frequency of loss of OCT1 activity worldwide. While most East Asian and Oceanian individuals had completely functional OCT1, 80 % of native South American Indians lacked functional OCT1 alleles. In East Asia and Oceania the average nucleotide diversity of the loss-of-function variants was much lower than that of the variants that do not affect OCT1 function (ratio of 0.03) and was significantly lower than the theoretically expected heterozygosity (Tajima's D = -1.64, P < 0.01). CONCLUSIONS: Comprehensive genetic analyses showed strong global variations in the frequency of loss of OCT1 activity with selection pressure for maintaining OCT1 activity in East Asia and Oceania. These results not only enable pharmacogenetically-based optimization of drug treatment worldwide, but may help elucidate the functional role of human OCT1.

10.
Breast Cancer Res Treat ; 137(1): 109-18, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23160925

RESUMO

Heparins seem to improve survival in patients with advanced malignancies independently of their anticoagulatory function. As the treatment options in advanced and metastatic breast cancer are still very limited, heparins might be an interesting addition to the existing systemic therapies. The interferon (IFN)-γ-inducible chemokines CXCL9 and CXCL10 play an essential role in the regulation of the immune milieu in malignant tumours, thereby being interesting targets for an immunological intervention. We therefore wanted to test whether heparins have an impact on the chemokines CXCL9 and CXCL10 as well as the IFN-γ signalling in human breast cancer cells in vitro. The well-established cell lines BT-474, MCF-7, SK-BR-3 and MDA-MB-231 were incubated with IFN-γ, unfractionated heparin (UFH), different low molecular weight heparins (LMWHs) and the heparin-related polyanions danaparoid and dextran sulphate. The production of CXCL9 and CXCL10 was measured by ELISA and real-time RT-PCR, the phosphorylation of signal transducer and activator of transcription (STAT) 1 was detected by an in-cell western assay and the amount of cellular bound IFN-γ was analysed by a high sensitivity ELISA. We observed that IFN-γ induced CXCL9 and CXCL10 production in MCF-7, SK-BR-3 and MDA-MB-231 cells but not in BT-474. UFH dose dependently inhibited the effect of IFN-γ on the secretion and expression of CXCL9 and CXCL10. LMWHs and heparin-related compounds differentially modulated IFN-γ-effects-the results depended on their molecular size and charge, but were independent of their anticoagulatory properties. As a reason for these heparin effects, we could show that the IFN-γ-induced phosphorylation of STAT1 was modulated by heparins, caused by an interaction with the cellular binding of IFN-γ. In conclusion, these results support the significance of the immunomodulatory properties of heparins independently of their classical anticoagulatory function. Heparin-derived sulphated polysaccharides with distinct molecular properties might thus be interesting candidates for new therapeutic strategies in breast cancer.


Assuntos
Antineoplásicos/farmacologia , Quimiocina CXCL10/metabolismo , Quimiocina CXCL9/metabolismo , Heparina/farmacologia , Interferon gama/fisiologia , Ativação Transcricional/efeitos dos fármacos , Linhagem Celular Tumoral , Quimiocina CXCL10/genética , Quimiocina CXCL9/genética , Feminino , Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Humanos , Interferon gama/antagonistas & inibidores , Fosforilação , Ligação Proteica , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Fator de Transcrição STAT1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...