Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Drug Metab Dispos ; 45(7): 770-778, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28428365

RESUMEN

Methamphetamine is one of the most abused illicit drugs with roughly 1.2 million users in the United States alone. A large portion of methamphetamine and its metabolites is eliminated by the kidney with renal clearance larger than glomerular filtration clearance. Yet the mechanism of active renal secretion is poorly understood. The goals of this study were to characterize the interaction of methamphetamine and its major metabolites with organic cation transporters (OCTs) and multidrug and toxin extrusion (MATE) transporters and to identify the major transporters involved in the disposition of methamphetamine and its major metabolites, amphetamine and para-hydroxymethamphetamine (p-OHMA). We used cell lines stably expressing relevant transporters to show that methamphetamine and its metabolites inhibit human OCTs 1-3 (hOCT1-3) and hMATE1/2-K with the greatest potencies against hOCT1 and hOCT2. Methamphetamine and amphetamine are substrates of hOCT2, hMATE1, and hMATE2-K, but not hOCT1 and hOCT3. p-OHMA is transported by hOCT1-3 and hMATE1, but not hMATE2-K. In contrast, organic anion transporters 1 and 3 do not interact with or transport these compounds. Methamphetamine and its metabolites exhibited complex interactions with hOCT1 and hOCT2, suggesting the existence of multiple binding sites. Our studies suggest the involvement of the renal OCT2/MATE pathway in tubular secretion of methamphetamine and its major metabolites and the potential of drug-drug interactions with substrates or inhibitors of the OCTs. This information may be considered when prescribing medications to suspected or known abusers of methamphetamine to mitigate the risk of increased toxicity or reduced therapeutic efficacy.


Asunto(s)
Transporte Biológico/fisiología , Metanfetamina/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Anfetamina/metabolismo , Sitios de Unión/fisiología , Línea Celular , Interacciones Farmacológicas/fisiología , Células HEK293 , Humanos , Riñón/metabolismo
2.
Xenobiotica ; 47(12): 1112-1120, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27977936

RESUMEN

1. Beta-carbolines are indole alkaloids with a wide range of pharmacological and toxicological activities. Beta-carbolines are structurally related to the neurotoxin 1-methyl-4-phenylpyridinium (MPP+), a known substrate of organic cation transporters (OCTs). The goal of this study is to determine the interaction of ß-carbolines with human OCT1, 2, and 3 (SLC22A1-3). 2. Dose-dependent inhibition studies were performed for five commercially available ß-carbolines using a fluorescent substrate assay in HEK293 cells stably expressing hOCT1-3. The substrate potential was evaluated by uptake assays and the impact of active transport on cellular toxicity examined. 3. All tested ß-carbolines potently inhibited hOCT2 with IC50 values in the sub- or low micromolar range. Harmaline is the most potent hOCT2 inhibitor (IC50 = 0.50 ± 0.08 µM). hOCT1 and hOCT3 are less sensitive to ß-carboline inhibition. Harmaline, norharmanium, and 2,9-dimethyl-4,9-dihydro-3H-ß-carbolinium accumulated 2- to 7-fold higher in cells expressing hOCT1-3. HEK293 cells expressing hOCT1-3 were 6.5- to 13-fold more sensitive to harmane and norharmanium toxicity. 4. Our data support a significant role of hOCT1-3 in tissue uptake and disposition of ß-carbolines. Importantly, the potent inhibition of hOCT2 by ß-carbolines also raises the concern of potential drug interactions between naturally occurring bioactive alkaloids and drugs eliminated by hOCT2.


Asunto(s)
Alcaloides/farmacología , Carbolinas/farmacología , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Transportador 2 de Cátion Orgánico
3.
J Pharmacol Exp Ther ; 359(3): 401-410, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27758931

RESUMEN

Renal transporter-mediated drug-drug interactions (DDIs) are of significant clinical concern, as they can adversely impact drug disposition, efficacy, and toxicity. Emerging evidence suggests that human renal organic cation transporter 2 (hOCT2) and multidrug and toxin extrusion proteins 1 and 2-K (hMATE1/2-K) exhibit substrate-dependent inhibition, but their impact on renal drug secretion and intracellular accumulation is unknown. Using metformin and atenolol as the probe substrates, we found that the classic inhibitors (e.g., cimetidine) of renal organic cation secretion were approximately 10-fold more potent for hOCT2 when atenolol was used, suggesting that atenolol is a more sensitive in vitro substrate for hOCT2 than metformin. In contrast, inhibition of hMATE1/2-K was influenced much less by the choice of substrate. Cimetidine is a much more potent inhibitor for hMATE1/2-K when metformin is the substrate but acts as an equally potent inhibitor of hOCT2 and hMATE1/2-K when atenolol is the substrate. Using hOCT2/hMATE1 double-transfected Madin-Darby canine kidney cells, we evaluated the impact of substrate-dependent inhibition on hOCT2/hMATE1-mediated transepithelial flux and intracellular drug accumulation. At clinically relevant concentrations, cimetidine dose dependently inhibited basal-to-apical flux of atenolol and metformin but impacted their intracellular accumulation differently, indicating that substrate-dependent inhibition may shift the major substrate-inhibitor interaction site between apical and basolateral transporters. Cimetidine is effective only when applied to the basal compartment. Our findings revealed the complex and dynamic nature of substrate-dependent inhibition of renal organic cation drug transporters and highlighted the importance of considering substrate-dependent inhibition in predicting transporter-mediated renal drug interaction, accumulation, and toxicity.


Asunto(s)
Interacciones Farmacológicas , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Riñón/citología , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Proteínas de Transporte de Catión Orgánico/metabolismo , Preparaciones Farmacéuticas/metabolismo , Animales , Atenolol/metabolismo , Atenolol/farmacología , Transporte Biológico/efectos de los fármacos , Cimetidina/metabolismo , Cimetidina/farmacología , Perros , Células HEK293 , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Células de Riñón Canino Madin Darby , Metformina/metabolismo , Metformina/farmacología , Transportador 2 de Cátion Orgánico , Preparaciones Farmacéuticas/química
4.
J Biol Chem ; 289(39): 27055-27064, 2014 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-25107910

RESUMEN

Drug-induced taste disturbance is a common adverse drug reaction often triggered by drug secretion into saliva. Very little is known regarding the molecular mechanisms underlying salivary gland transport of xenobiotics, and most drugs are assumed to enter saliva by passive diffusion. In this study, we demonstrate that salivary glands selectively and highly express OCT3 (organic cation transporter-3), a polyspecific drug transporter in the solute carrier 22 family. OCT3 protein is localized at both basolateral (blood-facing) and apical (saliva-facing) membranes of salivary gland acinar cells, suggesting a dual role of this transporter in mediating both epithelial uptake and efflux of organic cations in the secretory cells of salivary glands. Metformin, a widely used anti-diabetic drug known to induce taste disturbance, is transported by OCT3/Oct3 in vitro. In vivo, metformin was actively transported with a high level of accumulation in the salivary glands of wild-type mice. In contrast, active uptake and accumulation of metformin in salivary glands were abolished in Oct3(-/-) mice. Oct3(-/-) mice also showed altered metformin pharmacokinetics and reduced drug exposure in the heart. These results demonstrate that OCT3 is responsible for metformin accumulation and secretion in salivary glands. Our study uncovered a novel carrier-mediated pathway for drug entry into saliva and sheds new light on the molecular mechanisms underlying drug-induced taste disorders.


Asunto(s)
Hipoglucemiantes/farmacocinética , Metformina/farmacocinética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Glándulas Salivales/metabolismo , Animales , Transporte Biológico Activo/efectos de los fármacos , Transporte Biológico Activo/genética , Células HEK293 , Humanos , Hipoglucemiantes/efectos adversos , Hipoglucemiantes/farmacología , Metformina/efectos adversos , Metformina/farmacología , Ratones , Ratones Noqueados , Factor 3 de Transcripción de Unión a Octámeros/genética , Glándulas Salivales/patología , Trastornos del Gusto/inducido químicamente , Trastornos del Gusto/genética , Trastornos del Gusto/metabolismo , Trastornos del Gusto/patología
5.
Drug Metab Dispos ; 43(12): 1872-81, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26374172

RESUMEN

Atenolol is a ß-blocker widely used in the treatment of hypertension. Atenolol is cleared predominantly by the kidney by both glomerular filtration and active secretion, but the molecular mechanisms involved in its renal secretion are unclear. Using a panel of human embryonic kidney cell lines stably expressing human organic cation transporter (hOCT) 1-3, human organic anion transporter (hOAT) 1, hOAT3, human multidrug and toxin extrusion protein (hMATE) 1, and hMATE2-K, we found that atenolol interacted with both organic cation and anion transporters. However, it is transported by hOCT1, hOCT2, hMATE1, and hMATE2-K, but not by hOCT3, hOAT1, and hOAT3. A detailed kinetic analysis coupled with absolute quantification of membrane transporter proteins by liquid chromatography-tandem mass spectrometry revealed that atenolol is an excellent substrate for the renal transporters hOCT2, hMATE1, and hMATE2-K. The Km values for hOCT2, hMATE1, and hMATE2-K are 280 ± 4, 32 ± 5, and 76 ± 14 µM, respectively, and the calculated turnover numbers are 2.76, 0.41, and 2.20 s(-1), respectively. To demonstrate unidirectional transepithelial transport of atenolol, we developed and functionally validated a hOCT2/hMATE1 double-transfected Madin-Darby canine kidney cell culture model. Transwell studies showed that atenolol transport in the basal (B)-to-apical (A) direction is 27-fold higher than in the A-to-B direction, whereas its B-to-A/A-to-B transport ratio was only 2 in the vector-transfected control cells. The overall permeability of atenolol in the B-to-A direction in hOCT2/hMATE1 cells was 44-fold higher than in control cells. Together, our data support that atenolol tubular secretion is mediated through the hOCT2/hMATEs secretion pathway and suggest a significant role of organic cation transporters in the disposition of an important antihypertensive drug.


Asunto(s)
Atenolol/metabolismo , Riñón/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/fisiología , Proteínas de Transporte de Catión Orgánico/fisiología , Animales , Atenolol/farmacología , Perros , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Riñón/efectos de los fármacos , Células de Riñón Canino Madin Darby , Transportadores de Anión Orgánico Sodio-Independiente/antagonistas & inhibidores , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores
6.
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
7.
J Biol Chem ; 288(5): 3535-44, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23255610

RESUMEN

The choroid plexus (CP) forms the blood-cerebrospinal fluid (CSF) barrier and protects the brain from circulating metabolites, drugs, and toxins. The plasma membrane monoamine transporter (PMAT, SLC29A4) is a new polyspecific organic cation transporter that transports a wide variety of organic cations including biogenic amines, cationic drugs, and neurotoxins. PMAT is known to be expressed in the CP, but its specific role in CP transport of organic cations has not been clearly defined. Here we showed that PMAT transcript is highly expressed in human and mouse CPs, whereas transcripts of other functionally related transporters are minimally expressed in the CPs. Immunofluorescence staining further revealed that PMAT protein is localized to the apical (CSF-facing) membrane of the CP epithelium, consistent with a role of transporting organic cations from the CSF into CP epithelial cells. To further evaluate the role of PMAT in the CP, mice with targeted deletion of the Slc29a4 gene were generated and validated. Although Pmat(-/-) mice showed no overt abnormalities, the uptake of monoamines and the neurotoxin 1-methyl-4-phenylpyridinium was significantly reduced in CP tissues isolated from the knock-out mice. Together, our data demonstrated that PMAT is a major transporter for CP uptake of bioactive amines and xenobiotic cations. By removing its substrates from the CSF, PMAT may play an important role in protecting the brain from cationic neurotoxins and other potentially toxic organic cations.


Asunto(s)
Monoaminas Biogénicas/metabolismo , Membrana Celular/metabolismo , Plexo Coroideo/metabolismo , Marcación de Gen , Proteínas de Transporte de Catión Orgánico/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/genética , Animales , Cationes , Polaridad Celular/genética , Plexo Coroideo/citología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Proteínas de Transporte de Nucleósido Equilibrativas/metabolismo , Eliminación de Gen , Regulación de la Expresión Génica , Células HEK293 , Humanos , Concentración 50 Inhibidora , Ratones , Ratones Endogámicos C57BL , Transporte de Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/sangre , Proteínas de Transporte Vesicular de Monoaminas/metabolismo
8.
Biol Blood Marrow Transplant ; 20(10): 1544-52, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24923537

RESUMEN

Mycophenolic acid, the active metabolite of mycophenolate mofetil (MMF), inhibits inosine monophosphate dehydrogenase (IMPDH) activity. IMPDH is the rate-limiting enzyme involved in de novo synthesis of guanosine nucleotides and catalyzes the oxidation of inosine 5'-monophosphate to xanthosine 5'-monophosphate (XMP). We developed a highly sensitive liquid chromatography-mass spectrometry method to quantitate XMP concentrations in peripheral blood mononuclear cells (PMNCs) isolated from the recipient pretransplant and used this method to determine IMPDH activity in 86 nonmyeloablative allogeneic hematopoietic cell transplantation (HCT) patients. The incubation procedure and analytical method yielded acceptable within-sample and within-individual variability. Considerable between-individual variability was observed (12.2-fold). Low recipient pretransplant IMPDH activity was associated with increased day +28 donor T cell chimerism, more acute graft-versus-host disease (GVHD), lower neutrophil nadirs, and more cytomegalovirus reactivation but not with chronic GVHD, relapse, nonrelapse mortality, or overall mortality. We conclude that quantitation of the recipient's pretransplant IMPDH activity in PMNC lysate could provide a useful biomarker to evaluate a recipient's sensitivity to MMF. Further trials should be conducted to confirm our findings and to optimize postgrafting immunosuppression in nonmyeloablative HCT recipients.


Asunto(s)
Enfermedad Injerto contra Huésped/prevención & control , Neoplasias Hematológicas/terapia , Trasplante de Células Madre Hematopoyéticas , IMP Deshidrogenasa/metabolismo , Inmunosupresores/uso terapéutico , Leucocitos Mononucleares/enzimología , Ácido Micofenólico/análogos & derivados , Enfermedad Aguda , Adulto , Anciano , Biomarcadores/metabolismo , Femenino , Supervivencia de Injerto , Neoplasias Hematológicas/diagnóstico , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/mortalidad , Humanos , IMP Deshidrogenasa/antagonistas & inhibidores , Inosina Monofosfato/metabolismo , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/patología , Masculino , Persona de Mediana Edad , Ácido Micofenólico/uso terapéutico , Pronóstico , Estudios Prospectivos , Recurrencia , Ribonucleótidos/antagonistas & inhibidores , Ribonucleótidos/biosíntesis , Análisis de Supervivencia , Quimera por Trasplante/genética , Quimera por Trasplante/inmunología , Trasplante Homólogo , Xantina
9.
J Pharmacol Exp Ther ; 339(2): 376-85, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21816955

RESUMEN

Plasma membrane monoamine transporter (PMAT) is a new polyspecific transporter that interacts with a wide range of structurally diverse organic cations. To map the physicochemical descriptors of cationic compounds that allow interaction with PMAT, we systematically analyzed the interactions between PMAT and three series of structural analogs of known organic cation substrates including phenylalkylamines, n-tetraalkylammonium (n-TAA) compounds, and ß-carbolines. Our results showed that phenylalkylamines with a distance between the aromatic ring and the positively charged amine nitrogen atom of ∼6.4 Å confer optimal interactions with PMAT, whereas studies with n-TAA compounds revealed an excellent correlation between IC(50) values and hydrophobicity. The five ß-carbolines that we tested, which possess a pyridinium-like structure and are structurally related to the neurotoxin 1-methyl-4-phenylpyridinium, inhibited PMAT with high affinity (IC(50) values of 39.1-65.5 µM). Cytotoxicity analysis further showed that cells expressing PMAT are 14- to 15-fold more sensitive to harmalan and norharmanium, suggesting that these two ß-carbolines are also transportable substrates of PMAT. We then used computer-aided modeling to generate qualitative and quantitative three-dimensional pharmacophore models on the basis of 23 previously reported and currently identified PMAT inhibitors and noninhibitors. These models are characterized by a hydrogen bond donor and two to three hydrophobic features with distances between the hydrogen bond donor and hydrophobic features ranging between 5.20 and 7.02 Å. The consistency between the mapping results and observed PMAT affinity of a set of test compounds indicates that the models performed well in inhibitor prediction and could be useful for future virtual screening of new PMAT inhibitors.


Asunto(s)
Proteínas de Transporte de Nucleósido Equilibrativas/antagonistas & inhibidores , Proteínas de Transporte de Nucleósido Equilibrativas/química , Compuestos de Anilina/química , Compuestos de Anilina/metabolismo , Compuestos de Anilina/farmacología , Animales , Derivados del Benceno/química , Derivados del Benceno/metabolismo , Derivados del Benceno/farmacología , Carbolinas/química , Carbolinas/metabolismo , Carbolinas/farmacología , Línea Celular , Clonación Molecular , Perros , Evaluación Preclínica de Medicamentos , Proteínas de Transporte de Nucleósido Equilibrativas/genética , Proteínas de Transporte de Nucleósido Equilibrativas/metabolismo , Células HEK293 , Humanos , Concentración 50 Inhibidora , Modelos Moleculares , Conformación Molecular , Compuestos de Amonio Cuaternario/química , Compuestos de Amonio Cuaternario/metabolismo , Compuestos de Amonio Cuaternario/farmacología , Relación Estructura-Actividad
10.
J Pharmacol Exp Ther ; 335(3): 743-53, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20858707

RESUMEN

The plasma membrane monoamine transporter (PMAT) and organic cation transporter 3 (OCT3) are the two most prominent low-affinity, high-capacity (i.e., uptake(2)) transporters for endogenous biogenic amines. Using the Flp-in system, we expressed human PMAT (hPMAT) and human OCT3 (hOCT3) at similar levels in human embryonic kidney 293 cells. Parallel and detailed kinetics analysis revealed distinct and seemingly complementary patterns for the two transporters in transporting monoamine neurotransmitters. hPMAT is highly selective toward serotonin (5-HT) and dopamine, with the rank order of transport efficiency (V(max)/K(m)) being: dopamine, 5-HT ≫ histamine, norepinephrine, epinephrine. The substrate preference of hPMAT toward these amines is substantially driven by large (up to 15-fold) distinctions in its apparent binding affinities (K(m)). In contrast, hOCT3 is less selective than hPMAT toward the monoamines, and the V(max)/K(m) rank order for hOCT3 is: histamine > norepinephrine, epinephrine > dopamine >5-HT. It is noteworthy that hOCT3 demonstrated comparable (≤2-fold difference) K(m) toward all amines, and distinctions in V(max) played an important role in determining its differential transport efficiency toward the monoamines. Real-time reverse transcription-polymerase chain reaction revealed that hPMAT is expressed at much higher levels than hOCT3 in most human brain areas, whereas hOCT3 is selectively and highly expressed in adrenal gland and skeletal muscle. Our results suggest that hOCT3 represents a major uptake(2) transporter for histamine, epinephrine, and norepinephrine. hPMAT, on the other hand, is a major uptake(2) transporter for 5-HT and dopamine and may play a more important role in transporting these two neurotransmitters in the central nervous system.


Asunto(s)
Monoaminas Biogénicas/metabolismo , Transporte Biológico/fisiología , Proteínas de Transporte de Nucleósido Equilibrativas/metabolismo , Neurotransmisores/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , 1-Metil-4-fenilpiridinio/metabolismo , Glándulas Suprarrenales/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Encéfalo/metabolismo , ADN Nucleotidiltransferasas/genética , Dopamina/metabolismo , Epinefrina/metabolismo , Proteínas de Transporte de Nucleósido Equilibrativas/genética , Femenino , Expresión Génica/genética , Células HEK293 , Histamina/metabolismo , Humanos , Cinética , Masculino , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Endogámicos , Músculo Esquelético/metabolismo , Norepinefrina/metabolismo , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Proteínas de Transporte de Catión Orgánico/genética , Serotonina/metabolismo , Sinaptosomas/efectos de los fármacos , Sinaptosomas/metabolismo , Tetraetilamonio/metabolismo , Transfección
11.
Drug Metab Dispos ; 38(10): 1798-805, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20592246

RESUMEN

The plasma membrane monoamine transporter (PMAT) belongs to the equilibrative nucleoside transporter family (solute carrier 29) and was alternatively named equilibrative nucleoside transporter 4. Previous studies from our laboratory characterized PMAT as a polyspecific organic cation transporter that minimally interacts with nucleosides. Recently, PMAT-mediated uptake of adenosine (a purine nucleoside) was reported, and the transporter was proposed to function as a dual nucleoside/organic cation transporter. To clarify the substrate specificity of PMAT, we comprehensively analyzed the transport activity of human PMAT toward nucleosides, nucleobases, and organic cations in heterologous expression systems under well controlled conditions. Among 12 naturally occurring nucleosides and nucleobases, only adenosine was significantly transported by PMAT. PMAT-mediated adenosine transport is saturable, pH-dependent, and membrane-potential sensitive. Under both neutral (pH 7.4) and acidic (pH 6.6) conditions, adenosine is transported by PMAT at an efficiency (V(max)/K(m)) at least 10-fold lower than that of the organic cation substrates 1-methyl-4-phenylpyridinium and serotonin. PMAT-mediated adenosine uptake rate was significantly enhanced by an acidic extracellular pH. However, the effect of acidic pH was not adenosine-specific but was common to organic cation substrates as well. Our results demonstrated that although PMAT transports adenosine, the transporter kinetically prefers organic cation substrates. Functionally, PMAT should be viewed as a polyspecific organic cation transporter rather than an archetypical nucleoside transporter.


Asunto(s)
1-Metil-4-fenilpiridinio/metabolismo , Adenosina/metabolismo , Proteínas de Transporte de Nucleósido Equilibrativas/fisiología , Proteínas de Transporte de Catión Orgánico/fisiología , Serotonina/metabolismo , 1-Metil-4-fenilpiridinio/farmacocinética , Adenosina/farmacocinética , Animales , Transporte Biológico , Línea Celular , Relación Dosis-Respuesta a Droga , Proteínas de Transporte de Nucleósido Equilibrativas/genética , Proteínas de Transporte de Nucleósido Equilibrativas/metabolismo , Femenino , Concentración de Iones de Hidrógeno , Potenciales de la Membrana , Oocitos/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serotonina/farmacocinética , Especificidad por Sustrato , Transfección , Xenopus laevis
12.
Mol Endocrinol ; 23(4): 497-509, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19179481

RESUMEN

Star is expressed in steroidogenic cells as 3.5- and 1.6-kb transcripts that differ only in their 3'-untranslated regions (3'-UTR). In mouse MA10 testis and Y-1 adrenal lines, Br-cAMP preferentially stimulates 3.5-kb mRNA. ACTH is similarly selective in primary bovine adrenocortical cells. The 3.5-kb form harbors AU-rich elements (AURE) in the extended 3'-UTR, which enhance turnover. After peak stimulation of 3.5-kb mRNA, degradation is seen. Star mRNA turnover is enhanced by the zinc finger protein ZFP36L1/TIS11b, which binds to UAUUUAUU repeats in the extended 3'-UTR. TIS11b is rapidly stimulated in each cell type in parallel with Star mRNA. Cotransfection of TIS11b selectively decreases cytomegalovirus-promoted Star mRNA and luciferase-Star 3'-UTR reporters harboring the extended 3'-UTR. Direct complex formation was demonstrated between TIS11b and the extended 3'-UTR of the 3.5-kb Star. AURE mutations revealed that TIS11b-mediated destabilization required the first two UAUUUAUU motifs. HuR, which also binds AURE, did not affect Star expression. Targeted small interfering RNA knockdown of TIS11b specifically enhanced stimulation of 3.5-kb Star mRNA in bovine adrenocortical cells, MA-10, and Y-1 cells but did not affect the reversals seen after peak stimulation. Direct transfection of Star mRNA demonstrated that Br-cAMP stimulated a selective turnover of 3.5-kb mRNA independent of AURE, which may correspond to these reversal processes. Steroidogenic acute regulatory (STAR) protein induction was halved by TIS11b knockdown, concomitant with decreased cholesterol metabolism. TIS11b suppression of 3.5-kb mRNA is therefore surprisingly coupled to enhanced Star translation leading to increased cholesterol metabolism.


Asunto(s)
AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Procesamiento Postranscripcional del ARN , Proteínas de Unión al ARN/metabolismo , Dedos de Zinc , Regiones no Traducidas 3'/genética , Animales , Secuencia de Bases , Factor 1 de Respuesta al Butirato , Bovinos , Línea Celular , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Fosfoproteínas/genética , Estabilidad del ARN , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
13.
J Mol Endocrinol ; 38(1-2): 159-79, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17242178

RESUMEN

cAMP stimulation of rodent steroidogenic cells produces two StAR transcripts, a major 3.5 kb and a minor 1.6 kb mRNA, differing only in their 3' untranslated regions (3' UTR). They exhibit very different responses to stimulation and removal of 8-Br-cAMP, with the 3.5 kb form increasing and declining much more rapidly than the 1.6 kb form. The 3' end of the 3.5 kb StAR mRNA contains three conserved AU-rich element (AURE) motifs that mediate fast mRNA turnover in over 900 genes in the human genome. In this paper, we explore post-transcriptional regulation in steroidogenic and non-steroidogenic cells using expression vectors containing StAR or luciferase with different StAR 3' UTRs. We show that the basal steady-state levels of StAR or luciferase protein and mRNA are five to eight times lower with the 3' UTR of 3.5 kb StAR compared with that of the 1.6 kb 3' UTR. Examination of transcript stability by direct mRNA transfection showed only a 1.5-fold increase in the rate of cytoplasmic decay of the 3.5 kb mRNA relative to the 1.6 kb mRNA. However, the long 3' UTR caused a fivefold decrease in the rate of appearance of mature cytoplasmic mRNA despite transcription from the same promoter. This is attributed to less efficient nuclear processing of immature transcripts prior to export to cytoplasm. Selective 3' UTR sequence substitutions, deletions, and mutations showed that this loss of expression is produced additively by specific sequences in a 700-base basal instability region and by non-specific length effects. These mechanisms are selectively enhanced in steroidogenic cells. The AURE contribute a smaller basal destabilization effect selective for steroidogenic cells that is removed by their mutations. Inclusion of introns in the 3.5 kb StAR vector enhances StAR expression, suggesting the effects of introns complexes on nuclear processing. Br-cAMP provides an additional means to rapidly modulate StAR expression independent of transcription by attenuating the nuclear and cytoplasmic instability mechanisms within the extended 3' UTR.


Asunto(s)
Regiones no Traducidas 3'/genética , AMP Cíclico/fisiología , Inestabilidad Genómica , Fosfoproteínas/genética , ARN Mensajero/genética , Animales , Línea Celular Tumoral , Ratones , Ratas
14.
J Chem Neuroanat ; 83-84: 99-106, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27641077

RESUMEN

Plasma membrane monoamine transporter (PMAT) is a newly discovered monoamine transporter belonging to the equilibrative nucleoside transporter family. Highly expressed in the brain, PMAT represents a major uptake2 transporter that may play a role in monoamine clearance. Although human PMAT has been functionally characterized at the molecular level, rodent models are often used to evaluate PMAT function in ex vivo and in vivo studies. The aim of this study was to examine if there is potential species difference in the functional characteristics of PMAT between human, rat and mouse. A set of transfected cells stably expressing human PMAT (MDCK/hPMAT), rat Pmat (MDCK/rPmat) and mouse Pmat (Flp293/mPmat) were constructed. In MDCK/hPMAT, MDCK/rPmat and Flp293/mPmat cells, cellular localization analyses revealed that hPMAT, rPmat and mPmat are expressed and mainly localized to the plasma membranes of cells. The uptake of MPP+, serotonin and dopamine by MDCK/hPMAT, MDCK/rPmat and Flp293/mPmat cells was significantly increased compared with those by the mock transfection control. In contrast, two nucleosides, uridine and adenosine, minimally interacted with PMAT/Pmat in all species. The hPMAT-, rPmat- and mPmat-mediated uptakes of MPP+, serotonin and dopamine were saturable, with Km values of 33.7µM, 70.2µM and 49.5µM (MPP+), 116µM, 82.9µM and 231µM (serotonin), and 201µM, 271µM and 466µM (dopamine), respectively, suggesting similar substrate affinities between human and rodent PMAT/Pmat. The prototypical inhibitors, decynium 22 and GBR12935, also showed similar inhibition potencies between species. In conclusion, the present study demonstrated interspecies similarities in the functional characteristics of human and rodent PMAT/Pmat, which indicate a practical utility of rat and mouse animal models for further investigating and extrapolating the in vivo function of PMAT in humans.


Asunto(s)
Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/metabolismo , Animales , Humanos , Ratones , Ratas , Especificidad de la Especie
15.
Artículo en Inglés | MEDLINE | ID: mdl-27531991

RESUMEN

The cholesterol transfer function of steroidogenic acute regulatory protein (StAR) is uniquely integrated into adrenal cells, with mRNA translation and protein kinase A (PKA) phosphorylation occurring at the mitochondrial outer membrane (OMM). The StAR C-terminal cholesterol-binding domain (CBD) initiates mitochondrial intermembrane contacts to rapidly direct cholesterol to Cyp11a1 in the inner membrane (IMM). The conserved StAR N-terminal regulatory domain (NTD) includes a leader sequence targeting the CBD to OMM complexes that initiate cholesterol transfer. Here, we show how the NTD functions to enhance CBD activity delivers more efficiently from StAR mRNA in adrenal cells, and then how two factors hormonally restrain this process. NTD processing at two conserved sequence sites is selectively affected by StAR PKA phosphorylation. The CBD functions as a receptor to stimulate the OMM/IMM contacts that mediate transfer. The NTD controls the transit time that integrates extramitochondrial StAR effects on cholesterol homeostasis with other mitochondrial functions, including ATP generation, inter-organelle fusion, and the major permeability transition pore in partnership with other OMM proteins. PKA also rapidly induces two additional StAR modulators: salt-inducible kinase 1 (SIK1) and Znf36l1/Tis11b. Induced SIK1 attenuates the activity of CRTC2, a key mediator of StAR transcription and splicing, but only as cAMP levels decline. TIS11b inhibits translation and directs the endonuclease-mediated removal of the 3.5-kb StAR mRNA. Removal of either of these functions individually enhances cAMP-mediated induction of StAR. High-resolution fluorescence in situ hybridization (HR-FISH) of StAR RNA reveals asymmetric transcription at the gene locus and slow RNA splicing that delays mRNA formation, potentially to synchronize with cholesterol import. Adrenal cells may retain slow transcription to integrate with intermembrane NTD activation. HR-FISH resolves individual 3.5-kb StAR mRNA molecules via dual hybridization at the 3'- and 5'-ends and reveals an unexpectedly high frequency of 1:1 pairing with mitochondria marked by the matrix StAR protein. This pairing may be central to translation-coupled cholesterol transfer. Altogether, our results show that adrenal cells exhibit high-efficiency StAR activity that needs to integrate rapid cholesterol transfer with homeostasis and pulsatile hormonal stimulation. StAR NBD, the extended 3.5-kb mRNA, SIK1, and Tis11b play important roles.

16.
J Steroid Biochem Mol Biol ; 96(2): 155-73, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16039847

RESUMEN

The steroidogenic acute regulator (StAR) gene is transcribed to 1.6 kb and 3.5 kb mRNAs that differ only through the length of the 3'-untranslated region (3'-UTR). These forms result from alternative polyadenylation sites in exon 7. These sites are utilized similarly in unstimulated adrenal cells whereas Br-cAMP selectively stimulates 3.5 kb mRNA. After removal of Br-cAMP, 3.5 kb mRNA declines rapidly (t(1/2) = 2 h) while 1.6 kb mRNA responds more slowly. This selective degradation is more evident in testis MA10 cells and is seen even in the presence of Br-cAMP. Transfection of Y-1 cells with CMV promoted StAR vectors confirmed that the 3.5 kb form is less stable and that Br-cAMP slowly increases this instability. Basal instability resides solely in the extended 3'-UTR which contains AU-rich elements. Br-cAMP enhances this degradation of 3.5 kb mRNA but additionally requires translated and 5'-UTR sequences. Degradation of both forms is arrested by inhibitors of transcription or translation, indicating that mRNA stability is coupled to these processes independent of the extended 3'-UTR. Br-cAMP stimulates substantial selective synthesis of 3.5 kb StAR mRNA despite this instability. The preferential generation of the unstable form may facilitate rapid increases and decreases of StAR activity in response to external changes.


Asunto(s)
Regiones no Traducidas 3'/genética , Regulación de la Expresión Génica , Fosfoproteínas/genética , ARN Mensajero/genética , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Animales , Secuencia de Bases , Cartilla de ADN , Genes Reporteros , Humanos , Luciferasas/genética , Ratones , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Biosíntesis de Proteínas , ARN Mensajero/efectos de los fármacos , Ratas , Alineación de Secuencia , Homología de Secuencia de Ácido Nucleico , Transcripción Genética
17.
Mol Cell Endocrinol ; 336(1-2): 53-62, 2011 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-21147196

RESUMEN

The steroidogenic acute regulatory (StAR) protein is generated in rodents from 1.6 kb and 3.5 kb mRNA formed by alternative polyadenylation. The zinc finger protein, TIS11B (also Znf36L1), is elevated by cAMP in adrenal cells in parallel with StAR mRNA. TIS11b selectively destabilizes the 3.5 kb mRNA through AU-rich sequences at the end of the 3'UTR. siRNA suppression shows that TIS11b surprisingly increases StAR protein and cholesterol metabolism. StAR transcription is directly activated by PKA phosphorylation. cAMP responsive element binding (CREB) protein 1 phosphorylation is a key step leading to recruitment of the co-activator, CREB binding protein (CBP). A second protein, CREB regulated transcription coactivator (TORC/CRTC), enhances this recruitment, but is inhibited by salt inducible kinase (SIK). Basal StAR transcription is constrained through this phosphorylation of TORC. PKA provides an alternative stimulation by phosphorylating SIK, which prevents TORC inactivation. PKA stimulation of StAR nuclear transcripts substantially precedes TORC recruitment to the StAR promoter, which may, therefore, mediate a later step in mRNA production. Inhibition of SIK by staurosporine elevates StAR transcription and TORC recruitment to maximum levels, but without CREB phosphorylation. TORC suppression by SIK evidently limits basal StAR transcription. Staurosporine and cAMP stimulate synergistically. SIK targets the phosphatase, PP2a (activation), and Type 2 histone de-acetylases (inhibition), which may each contribute to suppression. Staurosporine stimulation through SIK inhibition is repeated in cAMP stimulation of many steroidogenic genes regulated by steroidogenic factor 1 (SF-1) and CREB. TIS11b and SIK may combine to attenuate StAR expression when hormonal stimuli decline.


Asunto(s)
Colesterol/metabolismo , AMP Cíclico/farmacología , Fosfoproteínas/metabolismo , ARN Mensajero/metabolismo , Transcripción Genética/efectos de los fármacos , Regiones no Traducidas 3'/genética , Animales , Humanos , Fosfoproteínas/genética , ARN Mensajero/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA