Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 300(9): 107629, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39098524

RESUMEN

Organic cations comprise a significant part of medically relevant drugs and endogenous substances. Such substances need organic cation transporters for efficient transfer via cell membranes. However, the membrane transporters of most natural or synthetic organic cations are still unknown. To identify these transporters, genes of 10 known OCTs and 18 orphan solute carriers (SLC) were overexpressed in HEK293 cells and characterized concerning their transport activities with a broad spectrum of low molecular weight substances emphasizing organic cations. Several SLC35 transporters and SLC38A10 significantly enhanced the transport of numerous relatively hydrophobic organic cations. Significant organic cation transport activities have been found in gene families classified as transporters of other substance classes. For instance, SLC35G3 and SLC38A10 significantly accelerated the uptake of several cations, such as clonidine, 3,4-methylenedioxymethamphetamine, and nicotine, which are known as substrates of a thus far genetically unidentified proton/organic cation antiporter. The transporters SLC35G4 and SLC35F5 stood out by their significantly increased choline uptake, and several other SLC transported choline together with a broader spectrum of organic cations. Overall, there are many more polyspecific organic cation transporters than previously estimated. Several transporters had one predominant substrate but accepted some other cationic substrates, and others showed no particular preference for one substrate but transported several organic cations. The role of these transporters in biology and drug therapy remains to be elucidated.


Asunto(s)
Proteínas de Transporte de Catión Orgánico , Humanos , Proteínas de Transporte de Catión Orgánico/metabolismo , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/química , Células HEK293 , Especificidad por Sustrato , Cationes/metabolismo , Transporte Biológico
2.
Int J Mol Sci ; 25(16)2024 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-39201429

RESUMEN

OCTN1 and OCTN2 are membrane transport proteins encoded by the SLC22A4 and SLC22A5 genes, respectively. Even though several transcripts have been predicted by bioinformatics for both genes, only one functional protein isoform has been described for each of them. Both proteins are ubiquitous, and depending on the physiopathological state of the cell, their expression is regulated by well-known transcription factors, although some aspects have been neglected. A plethora of missense variants with uncertain clinical significance are reported both in the dbSNP and the Catalogue of Somatic Mutations in Cancer (COSMIC) databases for both genes. Due to their involvement in human pathologies, such as inflammatory-based diseases (OCTN1/2), systemic primary carnitine deficiency (OCTN2), and drug disposition, it would be interesting to predict the impact of variants on human health from the perspective of precision medicine. Although the lack of a 3D structure for these two transport proteins hampers any speculation on the consequences of the polymorphisms, the already available 3D structures for other members of the SLC22 family may provide powerful tools to perform structure/function studies on WT and mutant proteins.


Asunto(s)
Regulación de la Expresión Génica , Miembro 5 de la Familia 22 de Transportadores de Solutos , Humanos , Miembro 5 de la Familia 22 de Transportadores de Solutos/genética , Miembro 5 de la Familia 22 de Transportadores de Solutos/metabolismo , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Proteínas de Transporte de Catión Orgánico/química , Conformación Proteica , Simportadores/genética , Simportadores/metabolismo , Simportadores/química
3.
Proc Natl Acad Sci U S A ; 117(9): 4732-4740, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32075917

RESUMEN

Multidrug and toxic compound extrusion (MATE) transporters are ubiquitous ion-coupled antiporters that extrude structurally and chemically dissimilar cytotoxic compounds and have been implicated in conferring multidrug resistance. Here, we integrate double electron-electron resonance (DEER) with functional assays and site-directed mutagenesis of conserved residues to illuminate principles of ligand-dependent alternating access of PfMATE, a proton-coupled MATE from the hyperthermophilic archaeon Pyrococcus furiosus Pairs of spin labels monitoring the two sides of the transporter reconstituted into nanodiscs reveal large-amplitude movement of helices that alter the orientation of a putative substrate binding cavity. We found that acidic pH favors formation of an inward-facing (IF) conformation, whereas elevated pH (>7) and the substrate rhodamine 6G stabilizes an outward-facing (OF) conformation. The lipid-dependent PfMATE isomerization between OF and IF conformation is driven by protonation of a previously unidentified intracellular glutamate residue that is critical for drug resistance. Our results can be framed in a mechanistic model of transport that addresses central aspects of ligand coupling and alternating access.


Asunto(s)
Antiportadores/química , Antiportadores/metabolismo , Proteínas de Transporte de Catión Orgánico/química , Proteínas de Transporte de Catión Orgánico/metabolismo , Antiportadores/genética , Resistencia a Múltiples Medicamentos , Espectroscopía de Resonancia por Spin del Electrón , Ligandos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Proteínas de Transporte de Catión Orgánico/genética , Conformación Proteica , Protones , Pyrococcus furiosus/metabolismo
4.
J Biol Chem ; 296: 100604, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33789160

RESUMEN

The membrane phospholipids phosphatidylcholine and phosphatidylethanolamine (PE) are synthesized de novo by the CDP-choline and CDP-ethanolamine (Kennedy) pathway, in which the extracellular substrates choline and ethanolamine are transported into the cell, phosphorylated, and coupled with diacylglycerol to form the final phospholipid product. Although multiple transport systems have been established for choline, ethanolamine transport is poorly characterized and there is no single protein assigned a transport function for ethanolamine. The solute carriers 44A (SLC44A) known as choline transporter-like proteins-1 and -2 (CTL1 and CTL2) are choline transporter at the plasma membrane and mitochondria. We report a novel function of CTL1 and CTL2 in ethanolamine transport. Using the lack or the gain of gene function in combination with specific antibodies and transport inhibitors we established two distinct ethanolamine transport systems of a high affinity, mediated by CTL1, and of a low affinity, mediated by CTL2. Both transporters are Na+-independent ethanolamine/H+ antiporters. Primary human fibroblasts with separate frameshift mutations in the CTL1 gene (M1= SLC44A1ΔAsp517 and M2= SLC44A1ΔSer126) are devoid of CTL1 ethanolamine transport but maintain unaffected CTL2 transport. The lack of CTL1 in M2 cells reduced the ethanolamine transport, the flux through the CDP-ethanolamine Kennedy pathway, and PE synthesis. In contrast, overexpression of CTL1 in M2 cells improved ethanolamine transport and PE synthesis. These data firmly establish that CTL1 and CTL2 are the first identified ethanolamine transporters in whole cells and mitochondria, with intrinsic roles in de novo PE synthesis by the Kennedy pathway and intracellular redistribution of ethanolamine.


Asunto(s)
Antígenos CD/metabolismo , Membrana Celular/metabolismo , Etanolamina/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Mitocondrias/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Animales , Antígenos CD/química , Transporte Biológico , Línea Celular , Humanos , Glicoproteínas de Membrana/química , Proteínas de Transporte de Membrana/química , Modelos Moleculares , Proteínas de Transporte de Catión Orgánico/química , Conformación Proteica
5.
J Biol Chem ; 296: 100262, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33837745

RESUMEN

In both prokaryotes and eukaryotes, multidrug and toxic-compound extrusion (MATE) transporters catalyze the efflux of a broad range of cytotoxic compounds, including human-made antibiotics and anticancer drugs. MATEs are secondary-active antiporters, i.e., their drug-efflux activity is coupled to, and powered by, the uptake of ions down a preexisting transmembrane electrochemical gradient. Key aspects of this mechanism, however, remain to be delineated, such as its ion specificity and stoichiometry. We previously revealed the existence of a Na+-binding site in a MATE transporter from Pyroccocus furiosus (PfMATE) and hypothesized that this site might be broadly conserved among prokaryotic MATEs. Here, we evaluate this hypothesis by analyzing VcmN and ClbM, which along with PfMATE are the only three prokaryotic MATEs whose molecular structures have been determined at atomic resolution, i.e. better than 3 Å. Reinterpretation of existing crystallographic data and molecular dynamics simulations indeed reveal an occupied Na+-binding site in the N-terminal lobe of both structures, analogous to that identified in PfMATE. We likewise find this site to be strongly selective against K+, suggesting it is mechanistically significant. Consistent with these computational results, DEER spectroscopy measurements for multiple doubly-spin-labeled VcmN constructs demonstrate Na+-dependent changes in protein conformation. The existence of this binding site in three MATE orthologs implicates Na+ in the ion-coupled drug-efflux mechanisms of this class of transporters. These results also imply that observations of H+-dependent activity likely stem either from a site elsewhere in the structure, or from H+ displacing Na+ under certain laboratory conditions, as has been noted for other Na+-driven transport systems.


Asunto(s)
Antiportadores/química , Proteínas de Transporte de Catión Orgánico/química , Conformación Proteica/efectos de los fármacos , Sodio/química , Antibacterianos/efectos adversos , Antibacterianos/farmacología , Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Antiportadores/ultraestructura , Sitios de Unión/efectos de los fármacos , Cristalografía por Rayos X , Humanos , Iones/química , Modelos Moleculares , Simulación de Dinámica Molecular , Proteínas de Transporte de Catión Orgánico/ultraestructura , Células Procariotas/química , Células Procariotas/ultraestructura , Dominios Proteicos/efectos de los fármacos
6.
PLoS Genet ; 15(9): e1008208, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31553721

RESUMEN

Variation in steroid hormone levels has wide implications for health and disease. The genes encoding the proteins involved in steroid disposition represent key determinants of interindividual variation in steroid levels and ultimately, their effects. Beginning with metabolomic data from genome-wide association studies (GWAS), we observed that genetic variants in the orphan transporter, SLC22A24 were significantly associated with levels of androsterone glucuronide and etiocholanolone glucuronide (sentinel SNPs p-value <1x10-30). In cells over-expressing human or various mammalian orthologs of SLC22A24, we showed that steroid conjugates and bile acids were substrates of the transporter. Phylogenetic, genomic, and transcriptomic analyses suggested that SLC22A24 has a specialized role in the kidney and appears to function in the reabsorption of organic anions, and in particular, anionic steroids. Phenome-wide analysis showed that functional variants of SLC22A24 are associated with human disease such as cardiovascular diseases and acne, which have been linked to dysregulated steroid metabolism. Collectively, these functional genomic studies reveal a previously uncharacterized protein involved in steroid homeostasis, opening up new possibilities for SLC22A24 as a pharmacological target for regulating steroid levels.


Asunto(s)
Proteínas de Transporte de Catión Orgánico/metabolismo , Esteroides/metabolismo , Simportadores/metabolismo , Androsterona/análogos & derivados , Androsterona/genética , Androsterona/metabolismo , Animales , Transporte Biológico , Estudio de Asociación del Genoma Completo/métodos , Células HEK293 , Humanos , Metabolómica/métodos , Modelos Moleculares , Proteínas de Transporte de Catión Orgánico/química , Proteínas de Transporte de Catión Orgánico/genética , Filogenia , Polimorfismo de Nucleótido Simple , Simportadores/química , Simportadores/genética
7.
Int J Mol Sci ; 23(2)2022 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-35055100

RESUMEN

The Novel Organic Cation Transporter, OCTN1, is the first member of the OCTN subfamily; it belongs to the wider Solute Carrier family SLC22, which counts many members including cation and anion organic transporters. The tertiary structure has not been resolved for any cation organic transporter. The functional role of OCNT1 is still not well assessed despite the many functional studies so far conducted. The lack of a definitive identification of OCTN1 function can be attributed to the different experimental systems and methodologies adopted for studying each of the proposed ligands. Apart from the contradictory data, the international scientific community agrees on a role of OCTN1 in protecting cells and tissues from oxidative and/or inflammatory damage. Moreover, the involvement of this transporter in drug interactions and delivery has been well clarified, even though the exact profile of the transported/interacting molecules is still somehow confusing. Therefore, OCTN1 continues to be a hot topic in terms of its functional role and structure. This review focuses on the most recent advances on OCTN1 in terms of functional aspects, physiological roles, substrate specificity, drug interactions, tissue expression, and relationships with pathology.


Asunto(s)
Biomarcadores , Susceptibilidad a Enfermedades , Interacciones Farmacológicas , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Simportadores/genética , Simportadores/metabolismo , Acetilación , Animales , Sitios de Unión , Transporte Biológico , Ergotioneína/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Modelos Moleculares , Conformación Molecular , Especificidad de Órganos , Proteínas de Transporte de Catión Orgánico/química , Unión Proteica , Relación Estructura-Actividad , Simportadores/química
8.
Biol Pharm Bull ; 44(4): 501-506, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33790101

RESUMEN

Multidrug and toxic compound extrusion (MATE) transporters are primarily expressed in the kidneys and liver, where they contribute to the excretion of organic cations. Our previous study suggested that pig MATE2 (class III) participates in testosterone secretion from Leydig cells. In humans, it is unclear which MATE class is involved in testosterone transport. In this study, we aimed to clarify whether human MATE1 (hMATE1) or human MATE2K (hMATE2K) mediates testosterone transport. To confirm that testosterone inhibits transporter-mediated tetraethylammonium (TEA) uptake, a cis-inhibition assay was performed using cells that stably expressed hMATE1 or hMATE2K. Docking simulations were performed to characterize differences in the binding of hMATE1 and hMATE2K to testosterone. Transport experiments in LLC-PK1 cells that stably expressed hMATE1 were used to test whether hMATE1 mediates testosterone transport. We detected differences between the amino acid sequences of the substrate-binding sites of hMATE1 and hMATE2K that could potentially be involved in testosterone binding. Testosterone and estradiol inhibited TEA uptake mediated by hMATE1 but not that mediated by hMATE2K. Transport experiments in LLC-PK1 cells indicated that testosterone might be transported via hMATE1. This study suggested that hMATE1, but not hMATE2K, is involved in human testosterone transport.


Asunto(s)
Proteínas de Transporte de Catión Orgánico/metabolismo , Testosterona/farmacología , Animales , Cimetidina/farmacología , Estradiol/farmacología , Células HEK293 , Humanos , Células LLC-PK1 , Modelos Moleculares , Proteínas de Transporte de Catión Orgánico/química , Porcinos , Tetraetilamonio/metabolismo
9.
Mol Divers ; 24(1): 141-154, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30868332

RESUMEN

hURAT1 (human urate transporter 1) is a successful target for hyperuricemia. Recently, the modification work on hURAT1 inhibitors showed that the flexible linkers would benefit biological activity. The study aimed to investigate the contribution of the linkers and give modification strategies on this kind of structures based on QSAR models (HQSAR and topomer CoMFA). The most effective HQSAR and topomer CoMFA models were generated by applying the training set containing 63 compounds, with the cross-validated q2 values of 0.869/0.818 and the non-cross-validated correlation coefficients r2 of 0.951/0.978, respectively. The Y-randomization test was applied to ensure the robustness of the models. The external predictive correlation coefficient (rpred2) grounded on the external test set (21 compounds) of two models was 0.910 and 0.907, respectively. In addition, the models were validated by Golbraikh-Tropsha and Roy methods, as well as other statistical metrics. The results showed that both models were reliable. Topomer CoMFA steric/electrostatic contours and HQSAR atomic contribution maps illustrated the structural features which governed their inhibitory potency. The dependable results could provide important insights to guide the designing of more potential hURAT1 inhibitors.


Asunto(s)
Descubrimiento de Drogas , Transportadores de Anión Orgánico/química , Proteínas de Transporte de Catión Orgánico/química , Relación Estructura-Actividad Cuantitativa , Algoritmos , Descubrimiento de Drogas/métodos , Humanos , Modelos Químicos , Modelos Moleculares , Conformación Molecular , Transportadores de Anión Orgánico/antagonistas & inhibidores , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores
10.
Allergy ; 74(11): 2167-2180, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31095759

RESUMEN

BACKGROUND: Despite all the efforts made up to now, the reasons that facilitate a protein becoming an allergen have not been elucidated yet. Alt a 1 protein is the major fungal allergen responsible for chronic asthma, but little is known about its immunological activity. Our main purpose was to investigate the ligand-dependent interactions of Alt a 1 in the human airway epithelium. METHODS: Alt a 1 with and without its ligand (holo- and apo- forms) was incubated with the pulmonary epithelial monolayer model, Calu-3 cells. Allergen transport and cytokine production were measured. Pull-down and immunofluorescence assays were employed to identify the receptor of Alt a 1 using the epithelial cell model and mouse tissues. Receptor-allergen-ligand interactions were analyzed by computational modeling. RESULTS: The holo-form could activate human monocytes, PBMCs, and polarized airway epithelial (Calu-3) cell lines. The allergen was also transported through the monolayer, without any alteration of the epithelial integrity (TEER). Alt a 1 also induced the production of proinflammatory IL8 and specific epithelial cytokines (IL33 and IL25) by Calu-3 cells. The interaction between epithelial cells and holo-Alt a 1 was found to be mediated by the SLC22A17 receptor, and its recognition of Alt a 1 was explained in structural terms. CONCLUSIONS: Our findings identified the Alt a 1 ligand as a central player in the interaction of the allergen with airway mucosa, shedding light into its potential role in the immunological response, while unveiling its potential as a new target for therapy intervention.


Asunto(s)
Antígenos Fúngicos/inmunología , Antígenos Fúngicos/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Presentación de Antígeno/inmunología , Antígenos Fúngicos/química , Biomarcadores , Línea Celular , Humanos , Leucocitos Mononucleares , Ligandos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Modelos Moleculares , Proteínas de Transporte de Catión Orgánico/química , Proteínas de Transporte de Catión Orgánico/genética , Unión Proteica , Conformación Proteica , Proteínas Recombinantes/inmunología , Relación Estructura-Actividad
11.
J Biol Chem ; 291(6): 2917-30, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26635366

RESUMEN

The neutrophil gelatinase-associated lipocalin (NGAL, also known as LCN2) and its cellular receptor (LCN2-R, SLC22A17) are involved in many physiological and pathological processes such as cell differentiation, apoptosis, and inflammation. These pleiotropic functions mainly rely on NGAL's siderophore-mediated iron transport properties. However, the molecular determinants underlying the interaction between NGAL and its cellular receptor remain largely unknown. Here, using solution-state biomolecular NMR in conjunction with other biophysical methods, we show that the N-terminal domain of LCN2-R is a soluble extracellular domain that is intrinsically disordered and interacts with NGAL preferentially in its apo state to form a fuzzy complex. The relatively weak affinity (≈10 µm) between human LCN2-R-NTD and apoNGAL suggests that the N terminus on its own cannot account for the internalization of NGAL by LCN2-R. However, human LCN2-R-NTD could be involved in the fine-tuning of the interaction between NGAL and its cellular receptor or in a biochemical mechanism allowing the receptor to discriminate between apo- and holo-NGAL.


Asunto(s)
Proteínas de Fase Aguda/química , Lipocalinas/química , Proteínas de Transporte de Catión Orgánico/química , Proteínas Proto-Oncogénicas/química , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Humanos , Lipocalina 2 , Lipocalinas/genética , Lipocalinas/metabolismo , Ratones , Resonancia Magnética Nuclear Biomolecular , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo
12.
Biochim Biophys Acta ; 1860(6): 1173-80, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26879959

RESUMEN

BACKGROUND: Multidrug and toxic compound extrusion (MATE) family transporters induce multiple-drug resistance (MDR) of bacterial pathogens and cancer cells, thus causing critical reductions in the therapeutic efficacies of antibiotics and anti-cancer drugs. Unfortunately, to date, the details and intrinsic reason about conformational regulation mechanism of MATE transporters remain elusive. METHOD: In this work, molecular dynamics (MD) simulations were conducted to explore the conformational regulation mechanism of PfMATE transporter from Pyrococcus furiosus based on different protonation state of Asp41. Two (MD) simulation systems were investigated: a system with protonation of Asp41 and a system without protonation of Asp41, which were named by D184(H)D41(H) system and D184(H) system, respectively. RESULTS AND CONCLUSIONS: Firstly, MD simulation results indicate that conformational changes mainly happen in extracellular regions of PfMATE protein. Further analysis reveals that PfMATE protein experiences different motion mode and forms different conformation based on different protonation state of Asp41. In the D184(H)D41(H) system, PfMATE experiences an opening motion and forms a more outward-open conformation. As for the D184(H) system, the protein has an anticlockwise rotational motion with the channel axis of protein and the more outward-open conformation does not appear. It can be inferred that protonation of Asp41 is essential for conformational regulation of PfMATE during transporting substrates. GENERAL SIGNIFICANCE: These findings provide intrinsic information for understanding the conformational regulation mechanism of PfMATE and will be very meaningful to explore the MDR mechanism of PfMATE further.


Asunto(s)
Simulación de Dinámica Molecular , Proteínas de Transporte de Catión Orgánico/química , Pyrococcus furiosus/metabolismo , Conformación Proteica
13.
Biochim Biophys Acta ; 1860(6): 1334-42, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26994919

RESUMEN

BACKGROUND: Organic cation transporters transfer solutes with a positive charge across the plasma membrane. The novel organic cation transporter 1 (OCTN1) and 2 (OCTN2) transport ergothioneine and carnitine, respectively. Mutations in the SLC22A5 gene encoding OCTN2 cause primary carnitine deficiency, a recessive disorders resulting in low carnitine levels and defective fatty acid oxidation. Variations in the SLC22A4 gene encoding OCTN1 are associated with rheumatoid arthritis and Crohn disease. METHODS: Here we evaluate the functional properties of the OCTN1 transporter using chimeric transporters constructed by fusing different portion of the OCTN1 and OCTN2 cDNAs. Their relative abundance and subcellular distribution was evaluated through western blot analysis and confocal microscopy. RESULTS: Substitutions of the C-terminal portion of OCTN1 with the correspondent residues of OCTN2 generated chimeric OCTN transporters more active than wild-type OCTN1 in transporting ergothioneine. Additional single amino acid substitutions introduced in chimeric OCTN transporters further increased ergothioneine transport activity. Kinetic analysis indicated that increased transport activity was due to an increased V(max), with modest changes in K(m) toward ergothioneine. CONCLUSIONS: Our results indicate that the OCTN1 transporter is tolerant to extensive amino acid substitutions. This is in sharp contrast to the OCTN2 carnitine transporter that has been selected for high functional activity through evolution, with almost all substitutions reducing carnitine transport activity. GENERAL SIGNIFICANCE: The widespread tolerance of OCTN1 to amino acid substitutions suggests that the corresponding SLC22A4 gene may have derived from a recent duplication of the SLC22A5 gene and might not yet have a defined physiological role.


Asunto(s)
Ergotioneína/farmacocinética , Proteínas de Transporte de Catión Orgánico/fisiología , Sustitución de Aminoácidos , Animales , Transporte Biológico , Western Blotting , Células CHO , Cricetulus , Humanos , Microscopía Confocal , Proteínas de Transporte de Catión Orgánico/química , Relación Estructura-Actividad , Simportadores
14.
J Biol Chem ; 290(46): 27633-43, 2015 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-26405039

RESUMEN

Organic cation transporter 3 (OCT3, SLC22A3) is a polyspecific, facilitative transporter expressed in astrocytes and in placental, intestinal, and blood-brain barrier epithelia, and thus elucidating the molecular mechanisms underlying OCT3 substrate recognition is critical for the rational design of drugs targeting these tissues. The pharmacology of OCT3 is distinct from that of other OCTs, and here we investigated the role of a hydrophobic cavity tucked within the translocation pathway in OCT3 transport properties. Replacement of an absolutely conserved Asp by charge reversal (D478E), neutralization (D478N), or even exchange (D478E) abolished MPP(+) uptake, demonstrating this residue to be obligatory for OCT3-mediated transport. Mutations at non-conserved residues lining the putative binding pocket of OCT3 to the corresponding residue in OCT1 (L166F, F450L, and E451Q) reduced the rate of MPP(+) transport, but recapitulated the higher sensitivity pharmacological profile of OCT1. Thus, interactions of natural polyamines (putrescine, spermidine, spermine) and polyamine-like potent OCT1 blockers (1,10-diaminodecane, decamethonium, bistriethylaminodecane, and 1,10-bisquinuclidinedecane) with wild-type OCT3 were weak, but were significantly potentiated in the mutant OCT3s. Conversely, a reciprocal mutation in OCT1 (F161L) shifted the polyamine-sensitivity phenotype toward that of OCT3. Further analysis indicated that OCT1 and OCT3 can recognize essentially the same substrates, but the strength of substrate-transporter interactions is weaker in OCT3, as informed by the distinct makeup of the hydrophobic cleft. The residues identified here are key contributors to both the observed differences between OCT3 and OCT1 and to the mechanisms of substrate recognition by OCTs in general.


Asunto(s)
Interacciones Hidrofóbicas e Hidrofílicas , Proteínas de Transporte de Catión Orgánico/química , Poliaminas/química , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Diaminas/química , Humanos , Datos de Secuencia Molecular , Mutación , Proteínas de Transporte de Catión Orgánico/genética , Transportador 1 de Catión Orgánico/química , Transportador 1 de Catión Orgánico/genética , Estructura Secundaria de Proteína , Putrescina/química , Ratas , Espermidina/química
15.
J Biol Chem ; 290(46): 27644-59, 2015 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-26432643

RESUMEN

The mitochondrial NAD pool is particularly important for the maintenance of vital cellular functions. Although at least in some fungi and plants, mitochondrial NAD is imported from the cytosol by carrier proteins, in mammals, the mechanism of how this organellar pool is generated has remained obscure. A transporter mediating NAD import into mammalian mitochondria has not been identified. In contrast, human recombinant NMNAT3 localizes to the mitochondrial matrix and is able to catalyze NAD(+) biosynthesis in vitro. However, whether the endogenous NMNAT3 protein is functionally effective at generating NAD(+) in mitochondria of intact human cells still remains to be demonstrated. To modulate mitochondrial NAD(+) content, we have expressed plant and yeast mitochondrial NAD(+) carriers in human cells and observed a profound increase in mitochondrial NAD(+). None of the closest human homologs of these carriers had any detectable effect on mitochondrial NAD(+) content. Surprisingly, constitutive redistribution of NAD(+) from the cytosol to the mitochondria by stable expression of the Arabidopsis thaliana mitochondrial NAD(+) transporter NDT2 in HEK293 cells resulted in dramatic growth retardation and a metabolic shift from oxidative phosphorylation to glycolysis, despite the elevated mitochondrial NAD(+) levels. These results suggest that a mitochondrial NAD(+) transporter, similar to the known one from A. thaliana, is likely absent and could even be harmful in human cells. We provide further support for the alternative possibility, namely intramitochondrial NAD(+) synthesis, by demonstrating the presence of endogenous NMNAT3 in the mitochondria of human cells.


Asunto(s)
Proteínas Portadoras/metabolismo , Citosol/metabolismo , Metaboloma , Mitocondrias/metabolismo , NAD/metabolismo , Secuencia de Aminoácidos , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Transporte Biológico , Proteínas Portadoras/química , Proteínas Portadoras/genética , Glucólisis , Células HEK293 , Humanos , Proteínas Mitocondriales , Datos de Secuencia Molecular , Nicotinamida-Nucleótido Adenililtransferasa/química , Nicotinamida-Nucleótido Adenililtransferasa/genética , Nicotinamida-Nucleótido Adenililtransferasa/metabolismo , Proteínas de Transporte de Nucleótidos , Proteínas de Transporte de Catión Orgánico/química , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
16.
Bioorg Med Chem Lett ; 26(2): 277-282, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26704267
17.
Arch Toxicol ; 90(7): 1555-84, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27165417

RESUMEN

Multidrug and toxin extrusion (MATE; SLC47A) proteins are membrane transporters mediating the excretion of organic cations and zwitterions into bile and urine and thereby contributing to the hepatic and renal elimination of many xenobiotics. Transported substrates include creatinine as endogenous substrate, the vitamin thiamine and a number of drug agents with in part chemically different structures such as the antidiabetic metformin, the antiviral agents acyclovir and ganciclovir as well as the antibiotics cephalexin and cephradine. This review summarizes current knowledge on the structural and molecular features of human MATE transporters including data on expression and localization in different tissues, important aspects on regulation and their functional role in drug transport. The role of genetic variation of MATE proteins for drug pharmacokinetics and drug response will be discussed with consequences for personalized medicine.


Asunto(s)
Quimioterapia , Proteínas de Transporte de Catión Orgánico/metabolismo , Medicina de Precisión , Procesamiento Proteico-Postraduccional , Secuencia de Aminoácidos , Animales , Biología Computacional , Humanos , Modelos Biológicos , Modelos Moleculares , Especificidad de Órganos , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Proteínas de Transporte de Catión Orgánico/química , Proteínas de Transporte de Catión Orgánico/genética , Preparaciones Farmacéuticas/química , Fosforilación , Alineación de Secuencia , Especificidad de la Especie , Especificidad por Sustrato
18.
Int J Mol Sci ; 17(5)2016 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-27144562

RESUMEN

Grain amaranth (Amaranthus hypochondriacus L.) is abundant in oxalate and can secrete oxalate under aluminium (Al) stress. However, the features of Al-induced secretion of organic acid anions (OA) and potential genes responsible for OA secretion are poorly understood. Here, Al-induced OA secretion in grain amaranth roots was characterized by ion charomatography and enzymology methods, and suppression subtractive hybridization (SSH) together with quantitative real-time PCR (qRT-PCR) was used to identify up-regulated genes that are potentially involved in OA secretion. The results showed that grain amaranth roots secrete both oxalate and citrate in response to Al stress. The secretion pattern, however, differs between oxalate and citrate. Neither lanthanum chloride (La) nor cadmium chloride (Cd) induced OA secretion. A total of 84 genes were identified as up-regulated by Al, in which six genes were considered as being potentially involved in OA secretion. The expression pattern of a gene belonging to multidrug and toxic compound extrusion (MATE) family, AhMATE1, was in close agreement with that of citrate secretion. The expression of a gene encoding tonoplast dicarboxylate transporter and four genes encoding ATP-binding cassette transporters was differentially regulated by Al stress, but the expression pattern was not correlated well with that of oxalate secretion. Our results not only reveal the secretion pattern of oxalate and citrate from grain amaranth roots under Al stress, but also provide some genetic information that will be useful for further characterization of genes involved in Al toxicity and tolerance mechanisms.


Asunto(s)
Aluminio/farmacología , Amaranthus/efectos de los fármacos , Ácidos Carboxílicos/metabolismo , Proteínas de Plantas/metabolismo , Amaranthus/metabolismo , Aniones/metabolismo , Ácido Cítrico/metabolismo , Proteínas de Transporte de Catión Orgánico/química , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Oxalatos/metabolismo , Proteínas de Plantas/química , Proteínas de Plantas/genética , Raíces de Plantas/efectos de los fármacos , Raíces de Plantas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Regulación hacia Arriba/efectos de los fármacos
19.
Biochim Biophys Acta ; 1838(10): 2539-46, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24967747

RESUMEN

MepA is a multidrug transporter from Staphylococcus aureus that confers multidrug resistance through the efflux of a wide array of hydrophobic substrates. To evaluate the ability of MepA to recognize different substrates, the dissociation constants for interactions between MepA and three of its substrates (acriflavine (Acr), rhodamine 6G (R6G), and ethidium (Et)) were measured. Given that MepA is purified in the presence of detergents and that its substrates are hydrophobic, we examined the effect of the detergent concentration on the dissociation constant. We demonstrate that all three substrates interact directly with the detergent micelles. Additionally, we find the detergent effect on the KD value to be highly substrate-dependent. The KD value for R6G is greatly influenced by the detergent, whereas the KD values for Acr and Et are only modestly affected. The effect of the inactive D183A mutant on binding was also evaluated. The D183A mutant shows lower affinity toward Acr and Et.


Asunto(s)
Proteínas Bacterianas/química , Farmacorresistencia Bacteriana Múltiple , Colorantes Fluorescentes/química , Proteínas de Transporte de Catión Orgánico/química , Staphylococcus aureus/química , Sustitución de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Detergentes/química , Colorantes Fluorescentes/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Micelas , Mutación Missense , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Unión Proteica , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Especificidad por Sustrato
20.
Biochim Biophys Acta ; 1828(2): 561-7, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23103507

RESUMEN

TBsmr is a secondary active multidrug transporter from Mycobacterium tuberculosis that transports a plethora of compounds including antibiotics and fluorescent dyes. It belongs to the small multidrug resistance (SMR) superfamily and is structurally and functionally related to E. coli EmrE. Of particular importance is the link between protein function, oligomeric state and lipid composition. By freeze fracture EM, we found three different size distributions in three different lipid environments for TBsmr indicating different oligomeric states. The link of these states with protein activity has been probed by fluorescence spectroscopy revealing significant differences. The drug binding site has been probed further by (19)F-MAS NMR through chemical labeling of native cysteine residues showing a water accessible environment in agreement with the alternating access model.


Asunto(s)
Antibacterianos/química , Antiportadores/química , Proteínas Bacterianas/química , Proteínas de Escherichia coli/metabolismo , Colorantes Fluorescentes/química , Lípidos/química , Mycobacterium tuberculosis/metabolismo , Proteínas de Transporte de Catión Orgánico/química , Antiportadores/metabolismo , Sitios de Unión , Cisteína/química , Escherichia coli/metabolismo , Técnica de Fractura por Congelación , Concentración de Iones de Hidrógeno , Espectroscopía de Resonancia Magnética/métodos , Microscopía Electrónica/métodos , Mutación , Espectrometría de Fluorescencia/métodos , Factores de Tiempo , Agua/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA