Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Haematologica ; 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38572568

RESUMEN

Belantamab mafodotin (belantamab) is a first-in-class anti-BCMA antibody-drug conjugate approved for the treatment of triple-class refractory multiple myeloma. It provides a unique therapeutic option for patients ineligible for CAR-T and bispecific antibody therapy, and/or patients progressing on anti-CD38 treatment where CAR-T and bispecifics might be kept in reserve. Wider use of the drug can be challenged by its distinct ocular side effect profile, including corneal microcysts and keratopathy. While dose reduction has been the most effective way to reduce these toxicities, the underlying mechanism of this BCMA off-target effect remains to be characterized. In this study, we provide the first evidence for soluble BCMA (sBCMA) in lacrimal fluid and report on its correlation with tumor burden in myeloma patients. We confirm that corneal cells do not express BCMA, and show that sBCMA-belantamab complexes may rather be internalized by corneal epithelial cells through receptor-ligand independent pinocytosis. Using an hTcEpi corneal cell-line model, we show that the pinocytosis inhibitor EIPA significantly reduces belantamab-specific cell killing. As a proof of concept, we provide detailed patient profiles demonstrating that, after belantamab-induced cell killing, sBCMA is released into circulation, followed by a delayed increase of sBCMA in the tear fluid and subsequent onset of keratopathy. Based on the proposed mechanism, pinocytosis-induced keratopathy can be prevented by lowering the entry of sBCMA into the lacrimal fluid. Future therapeutic concepts may therefore consist of belantamab-free debulking therapy prior to belantamab consolidation and/or concomitant use of gamma-secretase inhibition as currently evaluated for belantamab and nirogacestat in ongoing studies.

2.
Int J Biomed Imaging ; 2023: 4659041, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37484527

RESUMEN

The usefulness of perfluorocarbon nanoemulsions for the imaging of experimental myocarditis has been demonstrated in a high-field 9.4 Tesla MRI scanner. Our proof-of-concept study investigated the imaging capacity of PFC-based 19F/1H MRI in an animal myocarditis model using a clinical field strength of 1.5 Tesla. To induce experimental myocarditis, five male rats (weight ~300 g, age ~50 days) were treated with one application per week of doxorubicin (2 mg/kg BW) over a period of six weeks. Three control animals received the identical volume of sodium chloride 0.9% instead. Following week six, all animals received a single 4 ml injection of an 20% oil-in-water perfluorooctylbromide nanoemulsion 24 hours prior to in vivo1H/19F imaging on a 1.5 Tesla MRI. After euthanasia, cardiac histology and immunohistochemistry using CD68/ED1 macrophage antibodies were performed, measuring the inflamed myocardium in µm2 for further statistical analysis to compare the extent of the inflammation with the 19F-MRI signal intensity. All animals treated with doxorubicin showed a specific signal in the myocardium, while no myocardial signal could be detected in the control group. Additionally, the doxorubicin group showed a significantly higher SNR for 19F and a stronger CD68/ED1 immunhistoreactivity compared to the control group. This proof-of-concept study demonstrates that perfluorocarbon nanoemulsions could be detected in an in vivo experimental myocarditis model at a currently clinically relevant field strength.

3.
Adv Healthc Mater ; 12(13): e2201794, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36739269

RESUMEN

Nanohydrogels combine advantages of hydrogels and nanoparticles. In particular, they represent promising drug delivery systems. Nanogel synthesis by oxidative condensation of polyglycidol prepolymers, that are modified with thiol groups, results in crosslinking by disulfide bonds. Hereby, biomolecules like the antidiabetic peptide RS1-reg, derived from the regulatory protein RS1 of the Na+ -D-glucose cotransporter SGLT1, can be covalently bound by cysteine residues to the nanogel in a hydrophilic, stabilizing environment. After oral uptake, the acid-stable nanogels protect their loading during gastric passage from proteolytic degradation. Under alkaline conditions in small intestine the nanohydrogels become mucoadhesive, pass the intestinal mucosa and are taken up into small intestinal enterocytes by endocytosis. Using Caco-2 cells as a model for small intestinal enterocytes, by confocal laser scanning microscopy and structured illumination microscopy, the colocalization of fluorescent-labeled RS1-reg with markers of endosomes, lysosomes, and trans-Golgi-network after uptake with polyglycidol-based nanogels formed by precipitation polymerization is demonstrated. This indicates that RS1-reg follows the endosomal pathway. In the following, the design of bespoken nanohydrogels for specific targeting of RS1-reg to its site of action at the trans-Golgi network is described that might also represent a way of targeted transport for other drugs to their targets at the Golgi apparatus.


Asunto(s)
Proteínas de Transporte de Monosacáridos , Red trans-Golgi , Humanos , Nanogeles , Proteínas de Transporte de Monosacáridos/química , Proteínas de Transporte de Monosacáridos/metabolismo , Preparaciones Farmacéuticas , Células CACO-2 , Red trans-Golgi/metabolismo
4.
Adv Biol (Weinh) ; 6(10): e2101108, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35735188

RESUMEN

The regulatory solute carrier protein, family 1, member 1 (RS1) modulates via its N-terminal domain RS1-reg the activity of Na+ -d-glucose cotransporter 1 (SGLT1) and thereby the glucose uptake in the small intestine by blocking the release of SGLT1-containing vesicles at the trans-Golgi network (TGN). The antidiabetic activity of RS1 is mediated by ornithindecarboxylase 1 (ODC1), catalyzing the conversion of ornithine to putrescine. Putrescine can bind to a buddying protein complex for SGLT1-containing vesicles at the membrane of the TGN, triggering vesicle release. In this report, a first in-depth analysis of the important binding process between ODC1 and RS1-reg for regulating glucose uptake in the human organism is described by comparing results from the surface-based methods, "surface plasmon resonance" (SPR) and "surface acoustic wave" (SAW) with findings by isothermal titration calorimetry (ITC). In cases of SAW and SPR, three different assay surface setups are compared, resulting in small but significant differences in KD values for different surfaces. Noteworthy, an affinity increase by the factor of about 100 for the interaction is detected and herewith described for the first time in the presence of biological membranes that may be relevant in vivo for the biological function of RS1 and future bespoken antidiabetic drug development.


Asunto(s)
Proteínas de Transporte de Membrana Mitocondrial , Proteínas de Transporte de Monosacáridos , Putrescina , Humanos , Glucosa/metabolismo , Hipoglucemiantes , Proteínas de Transporte de Monosacáridos/metabolismo , Ornitina , Péptidos/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo
5.
Molecules ; 26(19)2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34641507

RESUMEN

Hyaluronic acid (HA)-based hydrogels are very commonly applied as cell carriers for different approaches in regenerative medicine. HA itself is a well-studied biomolecule that originates from the physiological extracellular matrix (ECM) of mammalians and, due to its acidic polysaccharide structure, offers many different possibilities for suitable chemical modifications which are necessary to control, for example, network formation. Most of these chemical modifications are performed using the free acid function of the polymer and, additionally, lead to an undesirable breakdown of the biopolymer's backbone. An alternative modification of the vicinal diol of the glucuronic acid is oxidation with sodium periodate to generate dialdehydes via a ring opening mechanism that can subsequently be further modified or crosslinked via Schiff base chemistry. Since this oxidation causes a structural destruction of the polysaccharide backbone, it was our intention to study a novel synthesis protocol frequently applied to selectively oxidize the C6 hydroxyl group of saccharides. On the basis of this TEMPO/TCC oxidation, we studied an alternative hydrogel platform based on oxidized HA crosslinked using adipic acid dihydrazide as the crosslinker.


Asunto(s)
Óxidos N-Cíclicos/química , Ácido Hialurónico/química , Hidrogeles/química , Células Madre Mesenquimatosas/citología , Adipatos/química , Diferenciación Celular , Supervivencia Celular , Condrogénesis , Reactivos de Enlaces Cruzados/química , Módulo de Elasticidad , Humanos , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Oxidación-Reducción , Bases de Schiff/química , Resonancia por Plasmón de Superficie
6.
Sensors (Basel) ; 21(9)2021 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-34062922

RESUMEN

Interactions between proteins and carbohydrates with larger biomacromolecules, e.g., lectins, are usually examined using self-assembled monolayers on target gold surfaces as a simplified model measuring setup. However, most of those measuring setups are either limited to a single substrate or do not allow for control over ligand distance and spacing. Here, we develop a synthetic strategy, consisting of a cascade of a thioesterification, native chemical ligation (NCL) and thiol-ene reaction, in order to create three-component polymer conjugates with a defined double bioactivation at the chain end. The target architecture is the vicinal attachment of two biomolecule residues to the α telechelic end point of a polymer and a thioether group at the ω chain end for fixating the conjugate to a gold sensor chip surface. As proof-of-principle studies for affinity measurements, we demonstrate the interaction between covalently bound mannose and ConA in surface acoustic wave (SAW) and surface plasmon resonance (SPR) experiments.


Asunto(s)
Oro , Oxazoles/química , Resonancia por Plasmón de Superficie , Concanavalina A , Lectinas , Manosa
7.
Mol Pharmacol ; 95(2): 169-182, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30409791

RESUMEN

Organic cation transporters OCT1 (SLC22A1) and OCT2 (SLC22A2) are critically involved in absorption and excretion of diverse cationic drugs. Because drug-drug interactions at these transporters may induce adverse drug effects in patients, in vitro testing during drug development for interaction with the human transporters is mandatory. Recent data performed with rat OCT1 (rOCT1) suggest that currently performed in vitro tests assuming one polyspecific binding site are insufficient. Here we measured the binding and transport of model substrate 1-methyl-4-phenylpyridinium+ (MPP+) by cell-free-expressed fusion proteins of rOCT1 and rOCT1 mutants with green fluorescent protein that had been reconstituted into nanodiscs or proteoliposomes. The nanodiscs were formed with major scaffold protein (MSP) and different phospholipids, whereas the proteoliposomes were formed with a mixture of cholesterol, phosphatidylserine, and phosphatidylcholine. In nanodiscs formed with 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine or cholesterol, phosphatidylserine, and phosphatidylcholine, two low-affinity MPP+ binding sites and one high-affinity MPP+ binding site per transporter monomer were determined. Mutagenesis revealed that tryptophan 218 and aspartate 475 in neighboring positions in the modeled outward-open cleft contribute to one low-affinity binding site, whereas arginine 440 located distantly in the cleft is critical for MPP+ binding to another low-affinity site. Comparing MPP+ binding with MPP+ transport suggests that the low-affinity sites are involved in MPP+ transport, whereas high-affinity MPP+ binding influences transport allosterically. The data will be helpful in the interpretation of future crystal structures and provides a rationale for future in vitro testing that is more sophisticated and reliable, leading to the generation of pharmacophore models with high predictive power.


Asunto(s)
1-Metil-4-fenilpiridinio/metabolismo , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Animales , Sitios de Unión , Proteínas Fluorescentes Verdes/metabolismo , Mutagénesis/fisiología , Fosfolípidos/metabolismo , Proteolípidos/metabolismo , Ratas
8.
Mol Pharmacol ; 95(1): 82-96, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30355744

RESUMEN

A domain of protein RS1 (RSC1A1) called RS1-Reg down-regulates the plasma membrane abundance of Na+-d-glucose cotransporter SGLT1 by blocking the exocytotic pathway at the trans-Golgi. This effect is blunted by intracellular glucose but prevails when serine in a QSP (Gln-Ser-Pro) motif is replaced by glutamate [RS1-Reg(S20E)]. RS1-Reg binds to ornithine decarboxylase (ODC) and inhibits ODC in a glucose-dependent manner. Because the ODC inhibitor difluoromethylornithine (DFMO) acts like RS1-Reg(S20E), and DFMO and RS1-Reg(S20E) are not cumulative, we raised the hypothesis that RS1-Reg(S20E) down-regulates the exocytotic pathway of SGLT1 at the trans-Golgi by inhibiting ODC. We investigated whether QEP down-regulates human SGLT1 (hSGLT1) like hRS1-Reg(S20E) and whether human Na+-d-glucose cotransporter hSGLT2 and the human glucose sensor hSGLT3 are also addressed. We expressed hSGLT1, hSGLT1 linked to yellow fluorescent protein (hSGLT1-YFP), hSGLT2-YFP and hSGLT3-YFP in oocytes of Xenopus laevis, injected hRS1-Reg(S20E), QEP, DFMO, and/or α-methyl-d-glucopyranoside (AMG), and measured AMG uptake, glucose-induced currents, and plasma membrane-associated fluorescence after 1 hour. We also performed in vitro AMG uptake measurements into small intestinal mucosa of mice and human. The data indicate that QEP down-regulates the exocytotic pathway of SGLT1 similar to hRS1-Reg(S20E). Our results suggests that both peptides also down-regulate hSGLT2 and hSGLT3 via the same pathway. Thirty minutes after application of 5 mM QEP in the presence of 5 mM d-glucose, hSGLT1-mediated AMG uptake into small intestinal mucosa was decreased by 40% to 50%. Thus oral application of QEP in a formulation that optimizes uptake into enterocytes but prevents entry into the blood is proposed as novel antidiabetic therapy.


Asunto(s)
Regulación hacia Abajo/fisiología , Exocitosis/fisiología , Glucosa/metabolismo , Proteínas de Transporte de Monosacáridos/metabolismo , Péptidos/metabolismo , Proteínas de Transporte de Sodio-Glucosa/metabolismo , Adulto , Animales , Transporte Biológico/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Eflornitina/farmacología , Exocitosis/efectos de los fármacos , Femenino , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Oocitos/metabolismo , Ornitina Descarboxilasa/metabolismo , Xenopus laevis
9.
Mol Pharmacol ; 90(5): 508-521, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27555600

RESUMEN

Na+-d-glucose cotransporter 1 (SGLT1) is rate-limiting for glucose absorption in the small intestine. Shortly after intake of glucose-rich food, SGLT1 abundance in the luminal membrane of the small intestine is increased. This upregulation occurs via glucose-induced acceleration of the release of SGLT1-containing vesicles from the trans-Golgi network (TGN), which is regulated by a domain of protein RS1 (RSC1A1) named RS1-Reg. Dependent on phosphorylation, RS1-Reg blocks release of vesicles containing SGLT1 or concentrative nucleoside transporter 1. The hypothesis has been raised that RS1-Reg binds to different receptor proteins at the TGN, which trigger release of vesicles with different transporters. To identify the presumed receptor proteins, two-hybrid screening was performed. Interaction with ornithine decarboxylase 1 (ODC1), the rate-limiting enzyme of polyamine synthesis, was observed and verified by immunoprecipitation. Binding of RS1-Reg mutants to ODC1 was characterized using surface plasmon resonance. Inhibition of ODC1 activity by RS1-Reg mutants and the ODC1 inhibitor difluoromethylornithine (DFMO) was measured in the absence and presence of glucose. In addition, short-term effects of DFMO, RS1-Reg mutants, the ODC1 product putrescine, and/or glucose on SGLT1 expressed in oocytes of Xenopus laevis were investigated. High-affinity binding of RS1-Reg to ODC1 was demonstrated, and evidence for a glucose binding site in ODC1 was provided. Binding of RS1-Reg to ODC1 inhibits the enzymatic activity at low intracellular glucose, which is blunted at high intracellular glucose. The data suggest that generation of putrescine by ODC1 at the TGN stimulates release of SGLT1-containing vesicles. This indicates a biomedically important role of ODC1 in regulation of glucose homeostasis.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Exocitosis/efectos de los fármacos , Glucosa/farmacología , Proteínas de Transporte de Monosacáridos/metabolismo , Ornitina Descarboxilasa/metabolismo , Transportador 1 de Sodio-Glucosa/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Células CACO-2 , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Eflornitina/farmacología , Electroforesis en Gel de Poliacrilamida , Células HEK293 , Humanos , Inmunoprecipitación , Espacio Intracelular/metabolismo , Cinética , Metilglucósidos/farmacología , Modelos Biológicos , Proteínas de Transporte de Monosacáridos/química , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Florizina/farmacología , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Proteínas Recombinantes/metabolismo , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Resonancia por Plasmón de Superficie , Xenopus laevis
10.
Front Pharmacol ; 7: 175, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27445813

RESUMEN

Lamivudine (3TC), a drug used in the treatment of HIV infection, needs to cross the plasma membrane to exert its therapeutic action. Human Organic cation transporter 1 (hOCT1), encoded by the SLC22A1 gene, is the transporter responsible for its uptake into target cells. As SLC22A1 is a highly polymorphic gene, the aim of this study was to determine how SNPs in the OCT1-encoding gene affected 3TC internalization and its interaction with other co-administered drugs. HEK293 cells stably transfected with either the wild type form or the polymorphic variants of hOCT1 were used to perform kinetic and drug-drug interaction studies. Protein co-immunoprecipitation was used to assess the impact of selected polymorphic cysteines on the oligomerization of the transporter. Results showed that 3TC transport efficiency was reduced in all polymorphic variants tested (R61C, C88R, S189L, M420del, and G465R). This was not caused by lack of oligomerization in case of variants located at the transporter extracellular loop (R61C and C88R). Drug-drug interaction measurements showed that co-administered drugs [abacavir (ABC), zidovudine (AZT), emtricitabine (FTC), tenofovir diproxil fumarate (TDF), efavirenz (EFV) and raltegravir (RAL)], differently inhibited 3TC uptake depending upon the polymorphic variant analyzed. These data highlight the need for accurate analysis of drug transporter polymorphic variants of clinical relevance, because polymorphisms can impact on substrate (3TC) translocation but even more importantly they can differentially affect drug-drug interactions at the transporter level.

11.
Mol Pharmacol ; 89(1): 118-32, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26464324

RESUMEN

Cellular uptake adapts rapidly to physiologic demands by changing transporter abundance in the plasma membrane. The human gene RSC1A1 codes for a 67-kDa protein named RS1 that has been shown to induce downregulation of the sodium-D-glucose cotransporter 1 (SGLT1) and of the concentrative nucleoside transporter 1 (CNT1) in the plasma membrane by blocking exocytosis at the Golgi. Injecting RS1 fragments into Xenopus laevis oocytes expressing SGLT1 or CNT1 and measuring the expressed uptake of α-methylglucoside or uridine 1 hour later, we identified a RS1 domain (RS1-Reg) containing multiple predicted phosphorylation sites that is responsible for this post-translational downregulation of SGLT1 and CNT1. Dependent on phosphorylation, RS1-Reg blocks the release of SGLT1-containing vesicles from the Golgi in a glucose-dependent manner or glucose-independent release of CNT1-containing vesicles. We showed that upregulation of SGLT1 in the small intestine after glucose ingestion is promoted by glucose-dependent disinhibition of the RS1-Reg-blocked exocytotic pathway of SGLT1 between meals. Mimicking phosphorylation of RS1-Reg, we obtained a RS1-Reg variant that downregulates SGLT1 in the brush-border membrane at high luminal glucose concentration. Because RS1 mediates short-term regulation of various transporters, we propose that the RS1-Reg-navigated transporter release from Golgi represents a basic regulatory mechanism of general importance, which implies the existence of receptor proteins that recognize different phosphorylated forms of RS1-Reg and of complex transporter-specific sorting in the trans-Golgi. RS1-Reg-derived peptides that downregulate SGLT1 at high intracellular glucose concentrations may be used for downregulation of glucose absorption in small intestine, which has been proposed as strategy for treatment of type 2 diabetes.


Asunto(s)
Exocitosis/fisiología , Glucosa/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Transporte de Monosacáridos/metabolismo , Transducción de Señal/fisiología , Transportador 1 de Sodio-Glucosa/metabolismo , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación/fisiología , Transportador 1 de Sodio-Glucosa/antagonistas & inhibidores , Xenopus laevis
12.
J Proteome Res ; 14(1): 353-65, 2015 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-25285821

RESUMEN

Improved procedures for sample preparation and proteomic data analysis allowed us to identify 7700 different proteins in mouse small intestinal mucosa and calculate the concentrations of >5000 proteins. We compared protein concentrations of small intestinal mucosa from mice that were fed for two months with normal diet (ND) containing 34.4% carbohydrates, 19.6% protein, and 3.3% fat or high-fat diet (HFD) containing 25.3% carbohydrates, 24.1% protein, and 34.6% fat. Eleven percent of the quantified proteins were significantly different between ND and HFD. After HFD, we observed an elevation of proteins involved in protein synthesis, protein N-glycosylation, and vesicle trafficking. Proteins engaged in fatty acid absorption, fatty acid ß-oxidation, and steroid metabolism were also increased. Enzymes of glycolysis and pentose phosphate cycle were decreased, whereas proteins of the respiratory chain and of ATP synthase were increased. The protein concentrations of various nutrient transporters located in the enterocyte plasma membrane including the Na(+)-d-glucose cotransporter SGLT1, the passive glucose transporter GLUT2, and the H(+)-peptide cotransporter PEPT1 were decreased. The concentration of the Na(+),K(+)-ATPase, which turned out to be the most strongly expressed enterocyte transporter, was also decreased. HFD also induced concentration changes of drug transporters and of enzymes involved in drug metabolism, which suggests effects of HFD on pharmacokinetics and toxicities. Finally, we observed down-regulation of antibody subunits and of components of the major histocompatibility complex II that may reflect impaired immune defense and immune tolerance in HFD. Our work shows dramatic changes in functional proteins of small intestine mucosa upon excessive fat consumption.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Metabolismo Energético/fisiología , Regulación de la Expresión Génica/fisiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Intestino Delgado , Proteoma/metabolismo , Animales , Proteínas Portadoras/metabolismo , Cromatografía Liquida , Ácidos Grasos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Proteoma/genética , Proteómica/métodos , Esteroides/metabolismo , Espectrometría de Masas en Tándem
13.
Mol Pharm ; 10(8): 3045-56, 2013 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-23763587

RESUMEN

Polyspecific organic cation transporter Oct2 from rat (gene Slc22A2) has been previously shown to transport Cs(+). Here we report that human OCT2 (hOCT2) is able to transport Cd(2+) showing substrate saturation with a Michaelis-Menten constant (Km) of 54 ± 5.8 µM. Uptake of Cd(2+) by hOCT2 was inhibited by typical hOCT2 ligands (unlabeled substrates and inhibitors), and the rate of uptake was decreased by a point mutation in a substrate binding domain of hOCT2. Incubation of hOCT2 overexpressing human embryonic kidney 293 cells (HEK-hOCT2-C) or rat renal proximal tubule cells expressing rOct2 (NRK-52E-C) with Cd(2+) resulted in an increased level of apoptosis that was reduced by OCT2 inhibitory ligand cimetidine(+). HEK-hOCT2-C exhibited different functional properties when they were confluent or had been dissociated by removal of Ca(2+) and Mg(2+). Only confluent HEK-hOCT2-C transported Cd(2+), and confluent and dissociated cells exhibited different potencies for inhibition of uptake of 1-methyl-4-phenylpyridinium(+) (MPP(+)) by Cd(2+), MPP(+), tetraethylammonium(+), cimetidine(+), and corticosterone. In confluent HEK-hOCT2-C, largely different inhibitor potencies were obtained upon comparison of inhibition of Cd(2+) uptake, 4-[4-(dimethylamino)styryl]-N-methylpyridinium(+) (ASP(+)) uptake, and MPP(+) uptake using substrate concentrations far below the respective Km values. Employing a point mutation in the previously identified substrate binding site of rat Oct1 produced evidence that short distance allosteric effects between binding sites for substrates and inhibitors are involved in substrate-dependent inhibitor potency. Substrate-dependent inhibitor affinity is probably a common property of OCTs. To predict interactions between drugs that are transported by OCTs and inhibitory drugs, it is necessary to employ the specific transported drug rather than a model substrate for in vitro measurements.


Asunto(s)
Cadmio/metabolismo , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Proteínas de Transporte de Catión Orgánico/metabolismo , Animales , Apoptosis/efectos de los fármacos , Cadmio/farmacología , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Línea Celular , Cimetidina/farmacología , Corticosterona/farmacología , Humanos , Túbulos Renales Proximales/metabolismo , Transportador 2 de Cátion Orgánico , Ratas , Tetraetilamonio/farmacología
14.
J Biol Chem ; 287(37): 31561-73, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22810231

RESUMEN

Organic cation transporters are membrane potential-dependent facilitative diffusion systems. Functional studies, extensive mutagenesis, and homology modeling indicate the following mechanism. A transporter conformation with a large outward-open cleft binds extracellular substrate, passes a state in which the substrate is occluded, turns to a conformation with an inward-open cleft, releases substrate, and subsequently turns back to the outward-open state. In the rat organic cation transporter (rOct1), voltage- and ligand-dependent movements of fluorescence-labeled cysteines were measured by voltage clamp fluorometry. For fluorescence detection, cysteine residues were introduced in extracellular parts of cleft-forming transmembrane α-helices (TMHs) 5, 8, and 11. Following expression of the mutants in Xenopus laevis oocytes, cysteines were labeled with tetramethylrhodamine-6-maleimide, and voltage-dependent conformational changes were monitored by voltage clamp fluorometry. One cysteine was introduced in the central domain of TMH 11 replacing glycine 478. This domain contains two amino acids that are involved in substrate binding and two glycine residues (Gly-477 and Gly-478) allowing for helix bending. Cys-478 could be modified with the transported substrate analog [2-(trimethylammonium)-ethyl]methanethiosulfonate but was inaccessible to tetramethylrhodamine-6-maleimide. Voltage-dependent movements at the indicator positions of TMHs 5, 8, and 11 were altered by substrate applications indicating large conformational changes during transport. The G478C exchange decreased transporter turnover and blocked voltage-dependent movements of TMHs 5 and 11. [2-(Trimethylammonium)-ethyl]methanethiosulfonate modification of Cys-478 blocked substrate binding, transport activity, and movement of TMH 8. The data suggest that Gly-478 is located within a mechanistically important hinge domain of TMH 11 in which substrate binding induces transport-related structural changes.


Asunto(s)
Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Activación del Canal Iónico/fisiología , Animales , Sitios de Unión , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/química , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/genética , Células HEK293 , Humanos , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Ratas , Especificidad por Sustrato/fisiología , Xenopus laevis
15.
J Biol Chem ; 286(43): 37874-86, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-21896487

RESUMEN

Polyspecific organic anion transporters (OATs) and organic cation transporters (OCTs) of the SLC22 transporter family play a pivotal role in absorption, distribution, and excretion of drugs. Polymorphisms in these transporters influence therapeutic effects. On the basis of functional characterizations, homology modeling, and mutagenesis, hypotheses for how OCTs bind and translocate structurally different cations were raised, assuming functionally competent monomers. However, homo-oligomerization has been described for OATs and OCTs. In the present study, evidence is provided that the large extracellular loops (EL) of rat Oct1 (rOct1) and rat Oat1 (rOat1) mediate homo- but not hetero-oligomerization. Replacement of the cysteine residues in the EL of rOct1 by serine residues (rOct1(6ΔC-l)) or breaking disulfide bonds with dithiothreitol prevented oligomerization. rOct1 chimera containing the EL of rOat1 (rOct1(rOat1-l)) showed oligomerization but reduced transporter amount in the plasma membrane. For rOct1(6ΔC-l) and rOct1(rOat1-l), similar K(m) values for 1-methyl-4-phenylpyridinium(+) (MPP(+)) and tetraethylammonium(+) (TEA(+)) were obtained that were higher compared with rOct1 wild type. The increased K(m) of rOct1(rOat1-l) indicates an allosteric effect of EL on the cation binding region. The similar substrate affinity of the oligomerizing and non-oligomerizing loop mutants suggests that oligomerization does not influence transport function. Independent transport function of rOct1 monomers was also demonstrated by showing that K(m) values for MPP(+) and TEA(+) were not changed after treatment with dithiothreitol and that a tandem protein with two rOct1 monomers showed about 50% activity with unchanged K(m) values for MPP(+) and TEA(+) when one monomer was blocked. The data help to understand how OCTs work and how mutations in patients may affect their functions.


Asunto(s)
Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Multimerización de Proteína/fisiología , Animales , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/química , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/genética , Ditiotreitol/química , Ditiotreitol/farmacología , Células HEK293 , Humanos , Transporte Iónico/efectos de los fármacos , Transporte Iónico/fisiología , Mutación , Proteína 1 de Transporte de Anión Orgánico/química , Proteína 1 de Transporte de Anión Orgánico/genética , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Multimerización de Proteína/efectos de los fármacos , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Ratas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Xenopus laevis
16.
Biochemistry ; 47(15): 4552-64, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18361503

RESUMEN

Polyspecific organic cation and anion transporters of the SLC22 protein family are critically involved in absorption and excretion of drugs. To elucidate transport mechanisms, functional and biophysical characterization of purified transporters is required and tertiary structures must be determined. Here, we synthesized rat organic cation transporters OCT1 and OCT2 and rat organic anion transporter OAT1 in a cell free system in the absence of detergent. We solubilized the precipitates with 2% 1-myristoyl-2-hydroxy- sn-glycero-3-[phospho- rac-(1-glycerol)] (LMPG), purified the transporters in the presence of 1% 3-[(3-cholamidopropyl)dimethylammonio]-1-propanesulfonate (CHAPS) or octyl glucoside, and reconstituted them into proteoliposomes. From 1 mL reaction vessels 0.13-0.36 mg of transporter proteins was purified. Thus, from five to ten 1 mL reaction vessels sufficient protein for crystallization was obtained. In the presence of 1% LMPG and 0.5% CHAPS, OCT1 and OAT1 formed homo-oligomers but no hetero-oligomers. After reconstitution of OCT1, OCT2, and OAT1 into proteoliposomes, similar Michaelis-Menten K m values were measured for uptake of 1-methyl-4-phenylpyridinium and p-aminohippurate (PAH (-)) by the organic cation and anion transporters, respectively, as after expression of the transporters in cells. Using the reconstituted system, evidence was obtained that OAT1 operates as obligatory and electroneutral PAH (-)/dicarboxylate antiporter and contains a low-affinity chloride binding site that stimulates turnover. PAH (-) uptake was observed only with alpha-ketoglutarate (KG (2-)) on the trans side, and trans-KG (2-) increased the PAH (-) concentration in voltage-clamped proteoliposomes transiently above equilibrium. The V max of PAH (-)/KG (2-) antiport was increased by Cl (-) in a manner independent of gradients, and PAH (-)/KG (2-) antiport was independent of membrane potential in the absence or presence of Cl (-).


Asunto(s)
Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , 1-Metil-4-fenilpiridinio/metabolismo , Animales , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/genética , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/aislamiento & purificación , Sistema Libre de Células , Ácidos Cetoglutáricos/metabolismo , Proteína 1 de Transporte de Anión Orgánico/genética , Proteína 1 de Transporte de Anión Orgánico/aislamiento & purificación , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/aislamiento & purificación , Transportador 2 de Cátion Orgánico , Proteolípidos/metabolismo , Ratas , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Ácido p-Aminohipúrico/metabolismo
17.
Am J Physiol Renal Physiol ; 293(3): F767-79, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17567940

RESUMEN

Effects of the sulfhydryl reagent methylmethanethiosulfonate (MMTS) on functions of organic cation transporters (OCTs) were investigated. Currents induced by 10 mM choline [I(max(choline))] in Xenopus laevis oocytes expressing rat OCT1 (rOCT1) were increased four- to ninefold after 30-s incubation with 5 mM MMTS whereas I(max(choline)) by rat OCT2 was 70% decreased. MMTS activated the rOCT1 transporter within the plasma membrane without changing stoichiometry between translocated charge and cation. After modification of oocytes expressing rOCT1 or rOCT2 with MMTS, I(0.5(choline)) values for choline-induced currents were increased. For rOCT1 it was shown that MMTS increased I(0.5) values for different cations by different degrees. Mutagenesis of individual cysteine residues in rOCT1 revealed that modification of cysteine 322 in the large intracellular loop, and of cysteine 451 at the transition of the transmembrane alpha-helix (TMH) 10 to the short intracellular loop between the TMH 10 and 11 is responsible for the observed effects of MMTS. After replacement of cysteine 451 by methionine, the IC(50(choline)) for choline to inhibit MPP uptake by rOCT1 was increased whereas the I(0.5(choline)) value for choline-induced current remained unchanged. At variance, in double mutant Cys322Ser, Cys451Met, I(0.5(choline)) was increased compared with rOCT1 wild-type whereas in the single mutant Cys322Ser I(0.5(choline)) was not changed. The data suggest that modification of rOCT1 at cysteines 322 and 451 leads to an increase in turnover. They indicate that cysteine 451 in rOCT1 interacts with the large intracellular loop and that cysteine 451 in both rOCT1 and rOCT2 is critical for the affinity of choline.


Asunto(s)
Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/genética , Cisteína/química , Proteínas de Transporte de Catión Orgánico/genética , Animales , Transporte Biológico Activo , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/química , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Capacidad Eléctrica , Electrofisiología , Exocitosis , Metilmetanosulfonato/farmacología , Modelos Moleculares , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Proteínas de Transporte de Catión Orgánico/química , Proteínas de Transporte de Catión Orgánico/metabolismo , Transportador 2 de Cátion Orgánico , Unión Proteica , Conformación Proteica , Ratas , Especificidad por Sustrato , Xenopus laevis
18.
Am J Physiol Renal Physiol ; 291(6): F1213-23, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16788146

RESUMEN

The product of gene RSC1A1, named RS1, participates in transcriptional and posttranscriptional regulation of the sodium-d-glucose cotransporter SGLT1. Using coexpression in oocytes of Xenopus laevis, posttranscriptional inhibition of human SGLT1 (hSGLT1) and some other transporters by human RS1 (hRS1) was demonstrated previously. In the present study, histidine-tagged hRS1 was expressed in oocytes or Sf9 cells and purified using nickel(II)-charged nitrilotriacetic acid-agarose. hRS1 protein was injected into oocytes expressing hSGLT1 or the human organic cation transporter hOCT2, and the effect on hSGLT1-mediated uptake of methyl-alpha-D-[14C]glucopyranoside ([14C]AMG) or hOCT2-mediated uptake of [14C]tetraethylammonium ([14C]TEA) was measured. Within 30 min after the injection of hRS1 protein, hSGLT1-expressed AMG uptake or hOCT2-expressed TEA uptake was inhibited by approximately 50%. Inhibition of AMG uptake was decreased when a dominant negative mutant of dynamin I was coexpressed and increased after stimulation of PKC. Inhibition remained unaltered when endocytosis was inhibited by chlorpromazine, imipramine, or filipin but was prevented when exocytosis was inhibited by botulinum toxin B or when the release of vesicles from the TGN and endosomes was inhibited by brefeldin A. Inhibition of hSGLT1-mediated AMG uptake and hOCT2-mediated TEA uptake by hRS1 protein were decreased at an enhanced intracellular AMG concentration. The data suggest that hRS1 protein exhibits glucose-dependent, short-term inhibition of hSGLT1 and hOCT2 by inhibiting the release of vesicles from the trans-Golgi network.


Asunto(s)
Exocitosis/fisiología , Proteínas de Transporte de Monosacáridos/metabolismo , Transportador 1 de Sodio-Glucosa/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Brefeldino A/farmacología , Radioisótopos de Carbono , Dinaminas/metabolismo , Exocitosis/efectos de los fármacos , Glucosa/metabolismo , Humanos , Insectos , Metilglucósidos/farmacocinética , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/farmacología , Oocitos/citología , Oocitos/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Transportador 2 de Cátion Orgánico , Proteína Quinasa C/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , Ratas , Sodio/metabolismo , Xenopus laevis , Red trans-Golgi/metabolismo
19.
Biochemistry ; 44(36): 12253-63, 2005 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-16142924

RESUMEN

The rat organic cation transporter rOCT1 with six histidine residues added to the C-terminus was expressed in Sf9 insect cells, and expression of organic cation transport was demonstrated. To purify rOCT1 protein, Sf9 cells were lysed with 1% (w/v) CHAPS [3-[(3-cholamidopropyl)dimethylammonio]-1-propanesulfonate], centrifuged, and subjected to sequential affinity chromatography using lentil-lectin Sepharose and nickel(II)-charged nitrilotriacetic acid-agarose. This procedure yielded approximately 70 microg of purified rOCT1 protein from 10 standard culture plates. Using a freeze-thaw procedure, purified rOCT1 was reconstituted into proteoliposomes formed from phosphatidylcholine, phosphatidylserine, and cholesterol. Proteoliposomes exhibited uptake of [3H]-1-Methyl-4-phenylpyridinium ([3H]MPP) that was inhibited by quinine and stimulated by an inside-negative membrane potential. MPP uptake was saturable with an apparent K(m) of 30 +/- 17 microM. MPP uptake (0.1 microM) was inhibited by tetraethylammonium, tetrabutylammonium, and tetrapentylammonium with IC50 values of 197 +/- 11, 19 +/- 1, and 1.8 +/- 0.03 microM, respectively. With membrane potential clamped to 0 mV using valinomycin in the presence of 100 mM potassium on both sides of the membrane, uptake of 0.1 microM MPP was trans stimulated 3-fold by 2.5 mM intracellular choline, and efflux of 0.1 microM MPP was trans stimulated 4-fold by 9.5 mM extracellular choline. The data show that rOCT1 is capable and sufficient to mediate transport of organic cations. The observed trans stimulation under voltage-clamp conditions shows that rOCT1 operates as a transporter rather than a channel. Purification and reconstitution of functional active rOCT1 protein is an important step toward the biophysical characterization and crystallization.


Asunto(s)
Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/aislamiento & purificación , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , 1-Metil-4-fenilpiridinio/farmacología , Animales , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/antagonistas & inhibidores , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/genética , Línea Celular , Expresión Génica , Proteolípidos/efectos de los fármacos , Proteolípidos/genética , Proteolípidos/metabolismo , Ratas , Spodoptera , Especificidad por Sustrato
20.
Blood Coagul Fibrinolysis ; 15(2): 169-78, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15091004

RESUMEN

Detection, purification, and partial characterization of a protease from Aeromonas hydrophila capable of cleaving prothrombin into active thrombin is described. The protease has been characterized with respect to enzymatic characteristics such as optimum reaction conditions for prothrombin activation, usage of additional substrates, as well as sensitivity against inhibitors. The protease activity can reversibly be inhibited by Me2+ chelating agents like ethylenediamine tetraacetic acid. The enzyme exhibits a pI value of 4.4 and can withstand temperatures up to 55 degrees C without loss of activity. With respect to prothrombin the enzyme exhibits a K(M) value of 1.47 micromol/l and a vmax value of 1.66 mol/min per mol enzyme. Amino terminal sequence analysis as well as mass spectrometry of fragments obtained by trypsin digest showed identity to a recently described elastase type protease from the same organism and homology to known proteases from other procaryotes (e.g. Aeromonas caviae, Vibrio proteolytica, Pseudomonas aeruginosa).


Asunto(s)
Aeromonas hydrophila/enzimología , Proteínas Bacterianas/farmacología , Metaloproteasas/farmacología , Protrombina/antagonistas & inhibidores , Clorometilcetonas de Aminoácidos/farmacología , Secuencia de Aminoácidos , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/metabolismo , Quelantes/farmacología , Ácido Edético/farmacología , Endopeptidasas/farmacología , Activación Enzimática/efectos de los fármacos , Punto Isoeléctrico , Metaloproteasas/aislamiento & purificación , Metaloproteasas/metabolismo , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Temperatura , Trombina/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA