Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Nature ; 595(7865): 130-134, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34040256

RESUMEN

Folates (also known as vitamin B9) have a critical role in cellular metabolism as the starting point in the synthesis of nucleic acids, amino acids and the universal methylating agent S-adenylsmethionine1,2. Folate deficiency is associated with a number of developmental, immune and neurological disorders3-5. Mammals cannot synthesize folates de novo; several systems have therefore evolved to take up folates from the diet and distribute them within the body3,6. The proton-coupled folate transporter (PCFT) (also known as SLC46A1) mediates folate uptake across the intestinal brush border membrane and the choroid plexus4,7, and is an important route for the delivery of antifolate drugs in cancer chemotherapy8-10. How PCFT recognizes folates or antifolate agents is currently unclear. Here we present cryo-electron microscopy structures of PCFT in a substrate-free state and in complex with a new-generation antifolate drug (pemetrexed). Our results provide a structural basis for understanding antifolate recognition and provide insights into the pH-regulated mechanism of folate transport mediated by PCFT.


Asunto(s)
Microscopía por Crioelectrón , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/metabolismo , Pemetrexed/química , Pemetrexed/metabolismo , Transportador de Folato Acoplado a Protón/química , Transportador de Folato Acoplado a Protón/metabolismo , Apoproteínas/química , Apoproteínas/metabolismo , Apoproteínas/ultraestructura , Transporte Biológico , Humanos , Modelos Moleculares , Transportador de Folato Acoplado a Protón/ultraestructura , Protones
2.
J Biol Chem ; 295(46): 15650-15661, 2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-32893190

RESUMEN

The proton-coupled folate transporter (PCFT, SLC46A1) is required for folate intestinal absorption and transport across the choroid plexus. Recent work has identified a F392V mutation causing hereditary folate malabsorption. However, the residue properties responsible for this loss of function remains unknown. Using site-directed mutagenesis, we observed complete loss of function with charged (Lys, Asp, and Glu) and polar (Thr, Ser, and Gln) Phe-392 substitutions and minimal function with some neutral substitutions; however, F392M retained full function. Using the substituted-cysteine accessibility method (with N-biotinyl aminoethyl methanethiosulfonate labeling), Phe-392 mutations causing loss of function, although preserving membrane expression and trafficking, also resulted in loss of accessibility of the substituted cysteine in P314C-PCFT located within the aqueous translocation pathway. F392V function and accessibility of the P314C cysteine were restored by insertion of a G305L (suppressor) mutation. A S196L mutation localized in proximity to Gly-305 by homology modeling was inactive. However, when inserted into the inactive F392V scaffold, function was restored (mutually compensatory mutations), as was accessibility of the P314C cysteine residue. Reduced function, documented with F392H PCFT, was due to a 15-fold decrease in methotrexate influx Vmax, accompanied by a decreased influx Kt (4.5-fold) and Ki (3-fold). The data indicate that Phe-392 is required for rapid oscillation of the carrier among its conformational states and suggest that this is achieved by dampening affinity of the protein for its folate substrates. F392V and other inactivating Phe-392 PCFT mutations lock the protein in its inward-open conformation. Reach (length) and hydrophobicity of Phe-392 appear to be features required for full activity.


Asunto(s)
Transportador de Folato Acoplado a Protón/metabolismo , Secuencia de Aminoácidos , Animales , Transporte Biológico , Cisteína/química , Cisteína/metabolismo , Deficiencia de Ácido Fólico/patología , Células HeLa , Humanos , Cinética , Síndromes de Malabsorción/patología , Metotrexato/metabolismo , Mutagénesis Sitio-Dirigida , Estructura Terciaria de Proteína , Transportador de Folato Acoplado a Protón/química , Transportador de Folato Acoplado a Protón/genética
3.
Proc Natl Acad Sci U S A ; 116(35): 17531-17540, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31405972

RESUMEN

Folates are critical for central nervous system function. Folate transport is mediated by 3 major pathways, reduced folate carrier (RFC), proton-coupled folate transporter (PCFT), and folate receptor alpha (FRα/Folr1), known to be regulated by ligand-activated nuclear receptors. Cerebral folate delivery primarily occurs at the choroid plexus through FRα and PCFT; inactivation of these transport systems can result in very low folate levels in the cerebrospinal fluid causing childhood neurodegenerative disorders. These disorders have devastating effects in young children, and current therapeutic approaches are not sufficiently effective. Our group has previously reported in vitro that functional expression of RFC at the blood-brain barrier (BBB) and its upregulation by the vitamin D nuclear receptor (VDR) could provide an alternative route for brain folate uptake. In this study, we further demonstrated in vivo, using Folr1 knockout (KO) mice, that loss of FRα led to a substantial decrease of folate delivery to the brain and that pretreatment of Folr1 KO mice with the VDR activating ligand, calcitriol (1,25-dihydroxyvitamin D3), resulted in over a 6-fold increase in [13C5]-5-formyltetrahydrofolate ([13C5]-5-formylTHF) concentration in brain tissues, with levels comparable to wild-type animals. Brain-to-plasma concentration ratio of [13C5]-5-formylTHF was also significantly higher in calcitriol-treated Folr1 KO mice (15-fold), indicating a remarkable enhancement in brain folate delivery. These findings demonstrate that augmenting RFC functional expression at the BBB could effectively compensate for the loss of Folr1-mediated folate uptake at the choroid plexus, providing a therapeutic approach for neurometabolic disorders caused by defective brain folate transport.


Asunto(s)
Encéfalo/metabolismo , Receptor 1 de Folato/metabolismo , Ácido Fólico/metabolismo , Proteína Portadora de Folato Reducido/metabolismo , Vitamina D/metabolismo , Animales , Transporte Biológico , Biomarcadores , Barrera Hematoencefálica/metabolismo , Cromatografía Liquida , Femenino , Receptor 1 de Folato/genética , Expresión Génica , Inmunohistoquímica , Ratones , Ratones Noqueados , Espectrometría de Masas en Tándem
4.
J Biol Chem ; 294(18): 7245-7258, 2019 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-30858177

RESUMEN

The proton-coupled folate transporter (PCFT) mediates intestinal absorption of folates and their transport from blood to cerebrospinal fluid across the choroid plexus. Substitutions at Asp-109 in the first intracellular loop between the first and second transmembrane domains (TMDs) abolish PCFT function, but protein expression and trafficking to the cell membrane are retained. Here, we used site-directed mutagenesis, the substituted-cysteine accessibility method, functional analyses, and homology modeling to determine whether the D109A substitution locks PCFT in one of its conformational states. Cys-substituted residues lining the PCFT aqueous translocation pathway and accessible in WT PCFT to the membrane-impermeable cysteine-biotinylation reagent, MTSEA-biotin, lost accessibility when introduced into the D109A scaffold. Substitutions at Gly-305 located exofacially within the eighth TMD, particularly with bulky residues, when introduced into the D109A scaffold largely restored function and MTSEA-biotin accessibility to Cys-substituted residues within the pathway. Likewise, Ser-196 substitution in the fifth TMD, predicted by homology modeling to be in proximity to Gly-305, also partially restored function found in solute transporters, is critical to oscillation of the carrier among its conformational states. Substitutions at Asp-109 and Gly-112 lock PCFT in an inward-open conformation, resulting in the loss of function. However, the integrity of the locked protein is preserved, indicated by the restoration of function after insertion of a second "unlocking" mutation. and accessibility. Similarly, the inactivating G112K substitution within the first intracellular loop was partially reactivated by introducing the G305L substitution. These data indicate that the first intracellular loop, with a sequence identical to "motif A" (GXXXDXXGR(R/K)).


Asunto(s)
Transportador de Folato Acoplado a Protón/metabolismo , Cisteína/química , Glicina/metabolismo , Células HeLa , Humanos , Cinética , Mutación , Conformación Proteica , Transportador de Folato Acoplado a Protón/química
5.
Mol Pharmacol ; 93(3): 208-215, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29326243

RESUMEN

The proton-coupled folate transporter (PCFT) is ubiquitously expressed in solid tumors to which it delivers antifolates, particularly pemetrexed, into cancer cells. Studies of PCFT-mediated transport, to date, have focused exclusively on the influx of folates and antifolates. This article addresses the impact of PCFT on concentrative transport, critical to the formation of the active polyglutamate congeners, and at pH levels relevant to the tumor microenvironment. An HeLa-derived cell line was employed, in which folate-specific transport was mediated exclusively by PCFT. At pH 7.0, there was a substantial chemical gradient for methotrexate that decreased as the extracellular pH was increased. A chemical gradient was still detected at pH 7.4 in the usual HEPES-based transport buffer in contrast to what was observed in a bicarbonate/CO2-buffered medium. This antifolate gradient correlated with an alkaline intracellular pH in the former (pH 7.85), but not the latter (pH 7.39), buffer and was abolished by the protonophore carbonyl cyanide-4-(trifluoromethoxy)phenylhydrazone. The gradient in HEPES buffer at pH 7.4 was the result of the activity of Na+/H+ exchanger(s); it was eliminated by inhibitors of Na+/H+ exchanger (s) or Na+/K+ ATPase. An antifolate chemical gradient was also detected in bicarbonate buffer at pH 6.9 versus 7.4, also suppressed by carbonyl cyanide-4-(trifluoromethoxy)phenylhydrazone. When the membrane potential is considered, PCFT generates substantial transmembrane electrochemical-potential gradients at extracellular pH levels relevant to the tumor microenvironment. The augmentation of intracellular pH, when cells are in a HEPES buffer, should be taken into consideration in studies that encompass all proton-coupled transporter families.


Asunto(s)
Antagonistas del Ácido Fólico/farmacocinética , Metotrexato/farmacocinética , Transportador de Folato Acoplado a Protón/metabolismo , Transporte Biológico Activo , Tampones (Química) , HEPES/farmacología , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Ácido Poliglutámico/metabolismo , Microambiente Tumoral
6.
Blood Adv ; 2(1): 61-68, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29344585

RESUMEN

Hereditary folate malabsorption (HFM) is an autosomal recessive disorder characterized by impaired intestinal folate absorption and impaired folate transport across the choroid plexus due to loss of function of the proton-coupled folate transporter (PCFT-SLC46A1). We report a novel mutation, causing HFM, affecting a residue located in the 11th transmembrane helix within the external gate. The mutant N411K-PCFT was stable, trafficked to the cell membrane, and had sufficient residual activity to characterize the transport defect and the structural requirements at this site for gate function. The influx Vmax of the N411K mutant was markedly decreased, as was the affinity for most, but not all, folate/antifolate substrates. The greatest loss of activity was for 5-methyltetrahydrofolate. Substitutions with positive charged residues resulted in a loss of activity (arginine > lysine > histidine). Function was retained for the negative charged aspartate, but not the larger glutamate substitutions, whereas the bulky hydrophobic (leucine), or polar (glutamine) substitutions, were tolerated. Homology models of PCFT, in the inward and outward open conformations, based upon the mammalian Glut5 fructose transporter structures, localize Asn411 protruding into the aqueous pathway. This is most prominent when the carrier is in the inward open conformation when the external gate is closed. Mutations at this site likely result in highly specific steric and electrostatic interactions between the Asn411-substituted, and other, residues in the gate region that impede carrier function. The substrate specificity of the N411K mutant may be due to alterations of substrate flows through the external gate, downstream allosteric alterations in the folate-binding pocket, or both.


Asunto(s)
Deficiencia de Ácido Fólico/genética , Síndromes de Malabsorción/genética , Mutación Missense , Transportador de Folato Acoplado a Protón/genética , Sustitución de Aminoácidos , Línea Celular , Deficiencia de Ácido Fólico/etiología , Células HeLa , Humanos , Lactante , Síndromes de Malabsorción/etiología , Masculino , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Transporte de Proteínas/genética , Especificidad por Sustrato , Tetrahidrofolatos/metabolismo , Transfección
7.
Am J Physiol Cell Physiol ; 314(3): C289-C296, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29167151

RESUMEN

The proton-coupled folate transporter (PCFT-SLC46A1) is required for folate transport across the apical membrane of the small intestine and across the choroid plexus. This study focuses on the structure/function of the 7th transmembrane domain (TMD), and its relationship to the 8th TMD as assessed by the substituted cysteine accessibility method (SCAM) and dicysteine cross-linking. Nine exofacial residues (I278C; H281C-L288C) of 23 residues in the 7th TMD were accessible to 2-((biotinoyl)amino)ethyl methanethiosulfonate (MTSEA-biotin). Pemetrexed, a high-affinity substrate for PCFT, decreased or abolished biotinylation of seven of these residues consistent with their location in or near the folate binding pocket. Homology models of PCFT based on Glut5 fructose transporter structures in both inward- and outward- open conformations were constructed and predicted that two pairs of residues (T289-I304C and Q285-Q311C) from the 7th and 8th TMDs should be in sufficiently close proximity to form a disulfide bond when substituted with cysteines. The single Cys-substituted mutants were accessible to MTSEA-biotin and functional with and without pretreatment with dithiotreitol. However, the double mutants were either not accessible at all, or accessibility was markedly reduced and function markedly impaired. This occurred spontaneously without inclusion of an oxidizing agent. Dithiotreitol restored accessibility and function consistent with disulfide bond disruption. The data establish the proximity of exofacial regions of the 7th and 8th TMDs and their role in defining the aqueous translocation pathway and suggest that these helices may be a component of an exofacial cleft through which substrates enter the protein binding pocket in its outward-open conformation.


Asunto(s)
Ácido Fólico/metabolismo , Transportador de Folato Acoplado a Protón/metabolismo , Transporte Biológico , Cisteína , Disulfuros/metabolismo , Células HeLa , Humanos , Cinética , Modelos Moleculares , Mutación , Oxidación-Reducción , Pemetrexed/metabolismo , Conformación Proteica en Hélice alfa , Dominios Proteicos , Transportador de Folato Acoplado a Protón/química , Transportador de Folato Acoplado a Protón/genética , Relación Estructura-Actividad
8.
Mol Pharm ; 14(11): 3848-3858, 2017 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-28885847

RESUMEN

Folates are essential for brain development and function. Folate transport in mammalian tissues is mediated by three major folate transport systems, i.e., reduced folate carrier (RFC), proton-coupled folate transporter (PCFT), and folate receptor alpha (FRα), known to be regulated by ligand-activated nuclear receptors, such as vitamin D receptor (VDR). Folate uptake at the choroid plexus, which requires the actions of both FRα and PCFT, is critical to cerebral folate delivery. Inactivating FRα or PCFT mutations cause severe cerebral folate deficiency resulting in early childhood neurodegeneration. The objective of this study was to investigate the role of RFC in folate uptake at the level of the blood-brain barrier (BBB) and its potential regulation by VDR. We detected robust expression of RFC in different in vitro BBB model systems, particularly in immortalized cultures of human cerebral microvascular endothelial cells (hCMEC/D3) and isolated mouse brain capillaries. [3H]-methotrexate uptake by hCMEC/D3 cells at pH 7.4 was inhibited by PT523 and pemetrexed, antifolates with high affinity for RFC. We also showed that activation of VDR through calcitriol (1,25-dihydroxyvitamin D3) exposure up-regulates RFC mRNA and protein expression as well as function in hCMEC/D3 cells and isolated mouse brain capillaries. We further demonstrated that RFC expression could be down-regulated by VDR-targeting siRNA, further confirming the role of VDR in the direct regulation of this folate transporter. Together, these data suggest that augmenting RFC functional expression could constitute a novel strategy for enhancing brain folate delivery for the treatment of neurometabolic disorders caused by loss of FRα or PCFT function.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Receptores de Calcitriol/metabolismo , Proteína Portadora de Folato Reducido/metabolismo , Animales , Transporte Biológico , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Calcitriol/farmacología , Células Cultivadas , Ácido Fólico/metabolismo , Humanos , Masculino , Metotrexato/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Wistar , Receptores de Calcitriol/genética , Proteína Portadora de Folato Reducido/genética
9.
Biochim Biophys Acta Biomembr ; 1859(11): 2193-2202, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28802835

RESUMEN

The proton-coupled folate transporter (PCFT-SLC46A1) is required for intestinal folate absorption and folate transport across the choroid plexus. This report addresses the structure/function of the 8th transmembrane helix. Based upon biotinylation of cysteine-substituted residues by MTSEA-biotin, 14 contiguous exofacial residues to Leu316 were accessible to the extracellular compartment of the 23 residues in this helix (Leu303-Leu325). Pemetrexed blocked biotinylation of six Cys-substituted residues deep within the helix implicating an important role for this region in folate binding. Accessibility decreased at 4°C vs RT. The influx Kt, Ki and Vmax were markedly increased for the P314C mutant, similar to what was observed for Y315A and Y315P mutants. However, the Kt, alone, was increased for the P314Y mutant. To correlate these observations with PCFT structural changes during the transport cycle, homology models were built for PCFT based upon the recently reported structures of bovine and rodent GLUT5 fructose transporters in the inward-open and outward- open conformations, respectively. The models predict substantial structural alterations in the exofacial region of the eighth transmembrane helix as it cycles between its conformational states that can account for the extended and contiguous aqueous accessibility of this region of the helix. Further, a helix break in one of the two conformations can account for the critical roles Pro314 and Tyr315, located in this region, play in PCFT function. The data indicates that the 8th transmembrane helix of PCFT plays an important role in defining the aqueous channel and the folate binding pocket.


Asunto(s)
Membrana Celular/química , Ácido Fólico/metabolismo , Dominios y Motivos de Interacción de Proteínas , Transportador de Folato Acoplado a Protón/química , Transportador de Folato Acoplado a Protón/metabolismo , Sustitución de Aminoácidos , Sitios de Unión/genética , Membrana Celular/metabolismo , Células HeLa , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Proteínas Mutantes/química , Unión Proteica/genética , Dominios Proteicos/genética , Dominios y Motivos de Interacción de Proteínas/genética , Transporte de Proteínas/genética , Transportador de Folato Acoplado a Protón/genética , Agua/química
10.
Am J Physiol Cell Physiol ; 312(4): C517-C526, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28122733

RESUMEN

The substituted cysteine accessibility method (SCAM) is widely used to study the structure and function of channels, receptors and transporters. In its usual application, a cysteine residue is introduced into a protein which lacks native cysteines following which the accessibility of the residue to the aqueous compartment is assessed. Implicit, and generally assumed, is that if the cysteine-substituted residue is not available to react with sulfhydryl reagents it is not exposed to the extracellular compartment or within the aqueous translocation pathway. We demonstrate here, in a Hela-derived cell line, that some cysteine-substituted residues of the proton-coupled folate transporter (PCFT, SLC46A1) that are inaccessible to 2-((biotinoyl)amino)ethyl methanethiosulfonate are glutathionylated by biotinylated glutathione ethyl ester in the absence of an oxidizing agent. Intramolecular disulfide formation involving cysteine-substituted residues was also identified in some instances. These posttranslational modifications limit the accessibility of the cysteine residues to sulfhydryl-reactive reagents and can have a profound impact on the interpretation of SCAM but may not alter function. When a posttranslationally modified residue is used as a reference extracellular control, the high level of exposure required for detection on Western blot results in erroneous detection of otherwise inaccessible intracellular cysteine-substituted residues. The data indicate that in the application of SCAM, when a cysteine-substituted residue does not appear to be accessible to sulfhydryl-reactive reagents, the possibility of a posttranslational modification should be excluded. The data explain the discrepancies in the assessment, and confirm the localization, of the first intracellular loop of PCFT.


Asunto(s)
Sustitución de Aminoácidos/fisiología , Cisteína/química , Cisteína/metabolismo , Glutatión/química , Glutatión/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Sitios de Unión , Células HeLa , Humanos , Unión Proteica , Ingeniería de Proteínas/métodos , Relación Estructura-Actividad
11.
Mol Aspects Med ; 53: 57-72, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27664775

RESUMEN

The proton-coupled folate transporter (PCFT-SLC46A1) is the mechanism by which folates are absorbed across the brush-border membrane of the small intestine. The transporter is also expressed in the choroid plexus and is required for transport of folates into the cerebrospinal fluid. Loss of PCFT function, as occurs in the autosomal recessive disorder "hereditary folate malabsorption" (HFM), results in a syndrome characterized by severe systemic and cerebral folate deficiency. Folate-receptor alpha (FRα) is expressed in the choroid plexus, and loss of function of this protein, as also occurs in an autosomal recessive disorder, results solely in "cerebral folate deficiency" (CFD), the designation for this disorder. This paper reviews the current understanding of the functional and structural properties and regulation of PCFT, an electrogenic proton symporter, and contrasts PCFT properties with those of the reduced folate carrier (RFC), an organic anion antiporter, that is the major route of folate transport to systemic tissues. The clinical characteristics of HFM and its treatment, based upon the thirty-seven known cases with the clinical syndrome, of which thirty have been verified by genotype, are presented. The ways in which PCFT and FRα might interact at the level of the choroid plexus such that each is required for folate transport from blood to cerebrospinal fluid are considered along with the different clinical presentations of HFM and CFD.


Asunto(s)
Deficiencia de Ácido Fólico/metabolismo , Síndromes de Malabsorción/metabolismo , Transportador de Folato Acoplado a Protón/metabolismo , Animales , Transporte Biológico , Deficiencia de Ácido Fólico/líquido cefalorraquídeo , Humanos , Absorción Intestinal , Síndromes de Malabsorción/líquido cefalorraquídeo , Modelos Moleculares , Transportador de Folato Acoplado a Protón/química
12.
Am J Physiol Cell Physiol ; 311(1): C150-7, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27251438

RESUMEN

The proton-coupled folate transporter (PCFT) mediates folate absorption across the brush-border membrane of the proximal small intestine and is required for folate transport across the choroid plexus into the cerebrospinal fluid. In this study, the functional role and accessibility of the seven PCFT Trp residues were assessed by the substituted-cysteine accessibility method. Six Trp residues at a lipid-aqueous interface tolerated Cys substitution in terms of protein stability and function. W85C, W202C, and W213C were accessible to N-biotinyl aminoethylmethanethiosulfonate; W48C and W299C were accessible only after treatment with dithiotreitol (DTT), consistent with modification of these residues by an endogenous thiol-reacting molecule and their extracellular location. Neither W107C nor W333C was accessible (even after DTT) consistent with their cytoplasmic orientation. Biotinylation was blocked by pemetrexed only for the W48C (after DTT), W85C, W202C residues. Function was impaired only for the W299C PCFT mutant located in the 4th external loop between the 7th and 8th transmembrane helices. Despite its aqueous location, function could only be fully preserved with Phe and, to a lesser extent, Ala substitutions. There was a 6.5-fold decrease in the pemetrexed influx Vmax and a 3.5- and 6-fold decrease in the influx Kt and Ki, respectively, for the W299S PCFT. The data indicate that the hydrophobicity of the W299 residue is important for function suggesting that during the transport cycle this residue interacts with the lipid membrane thereby impacting on the oscillation of the carrier and, indirectly, on the folate binding pocket.


Asunto(s)
Membrana Celular/metabolismo , Ácido Fólico/metabolismo , Transportador de Folato Acoplado a Protón/metabolismo , Sitios de Unión , Biotinilación , Membrana Celular/efectos de los fármacos , Cisteína , Antagonistas del Ácido Fólico/farmacología , Genotipo , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Mutagénesis Sitio-Dirigida , Mutación , Pemetrexed/farmacología , Fenotipo , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios Proteicos , Estabilidad Proteica , Transportador de Folato Acoplado a Protón/química , Transportador de Folato Acoplado a Protón/efectos de los fármacos , Transportador de Folato Acoplado a Protón/genética , Relación Estructura-Actividad , Transfección , Triptófano
13.
J Biol Chem ; 291(15): 8162-72, 2016 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-26884338

RESUMEN

The proton-coupled folate transporter (PCFT, SLC46A1) is required for intestinal folate absorption and folate homeostasis in humans. A homology model of PCFT, based upon theEscherichia coliglycerol 3-phosphate transporter structure, predicted that PCFT transmembrane domains (TMDs) 1, 2, 7, and 11 form an extracellular gate in the inward-open conformation. To assess this model, five residues (Gln(45)-TMD1, Asn(90)-TMD2, Leu(290)-TMD7, Ser(407)-TMD11 and Asn(411)-TMD11) in the predicted gate were substituted with Cys to generate single and nine double mutants. Transport function of the mutants was assayed in transient transfectants by measurement of [(3)H]substrate influx as was accessibility of the Cys residues to biotinylation. Pairs of Cys residues were assessed for spontaneous formation of a disulfide bond, induction of a disulfide bond by oxidization with dichloro(1,10-phenanthroline)copper (II) (CuPh), or the formation of a Cd(2+)complex. The data were consistent with the formation of a spontaneous disulfide bond between the N90C/S407C pair and a CuPh- and Cd(2+)-induced disulfide bond and complex, respectively, for the Q45C/L290C and L290C/N411C pairs. The decrease in activity induced by cross-linkage of the Cys residue pairs was due to a decrease in the influxVmaxconsistent with restriction in the mobility of the transporter. The presence of folate substrate decreased the CuPh-induced inhibition of transport. Hence, the data support the glycerol 3-phosphate transporter-based homology model of PCFT and the presence of an extracellular gate formed by TMDs 1, 2, 7, and 11.


Asunto(s)
Cisteína/química , Disulfuros/química , Ácido Fólico/metabolismo , Transportador de Folato Acoplado a Protón/química , Transportador de Folato Acoplado a Protón/metabolismo , Sustitución de Aminoácidos , Cisteína/genética , Cisteína/metabolismo , Disulfuros/metabolismo , Células HeLa , Humanos , Modelos Moleculares , Mutación , Oxidación-Reducción , Conformación Proteica , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Transportador de Folato Acoplado a Protón/genética
14.
Cancer Chemother Pharmacol ; 75(6): 1163-73, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25847479

RESUMEN

PURPOSE: Elements in the endocytic process that are determinants of the activities of antifolates delivered by folate-receptor alpha (FRα) were explored. METHODS: Antifolate growth inhibition was assessed with a 1- or 5-day exposure in reduced folate carrier-null HeLa cell lines that express a high level of FRα in the presence or absence of the proton-coupled folate transporter (PCFT). pH-dependent rates of dissociation from FRα were also determined. RESULTS: With a 1-day drug exposure which is relevant to the pulse clinical administration of these drugs, FRα expression enhanced raltitrexed activity and modestly enhanced ZD9331 activity, but did not significantly augment the activity of pemetrexed or lomotrexol. With a 5-day drug exposure, FRα-mediated growth inhibition was increased for raltitrexed and ZD9331 and emerged for lomotrexol. While the FRα-augmented activity of lomotrexol and raltitrexed did not require PCFT, augmentation of ZD9331 activity required the co-expression of PCFT with both 1- and 5-day exposures. In contrast, there was no augmentation of pemetrexed activity by FRα under any condition. The activities of these agents correlated with their rate of dissociation from the receptor at acidic pH: raltitrexed > ZD9331 > lomotrexol > pemetrexed consistent with insufficient pemetrexed release from FRα for export from the endosomes. CONCLUSIONS: FRα is unlikely to contribute to the pharmacological activity of antifolates, such as pemetrexed, that bind tightly to, and dissociate slowly from, the receptor particularly when the exposure time is brief. While PCFT was required for FRα-mediated ZD9931 activity, the activities of the other antifolates was independent of PCFT.


Asunto(s)
Endocitosis/fisiología , Receptor 1 de Folato/metabolismo , Antagonistas del Ácido Fólico/farmacología , Ácido Fólico/metabolismo , Transportador de Folato Acoplado a Protón/metabolismo , Línea Celular Tumoral , Glutamatos/farmacología , Guanina/análogos & derivados , Guanina/farmacología , Células HeLa , Humanos , Pemetrexed , Quinazolinas/farmacología , Proteína Portadora de Folato Reducido/metabolismo , Tiofenos/farmacología
15.
Artículo en Inglés | MEDLINE | ID: mdl-25562642

RESUMEN

Susan Band Horwitz is a Distinguished Professor and holds the Falkenstein Chair in Cancer Research at Albert Einstein College of Medicine in New York. She is co-chair of the Department of Molecular Pharmacology and associate director for therapeutics at the Albert Einstein Cancer Center. After graduating from Bryn Mawr College, Dr. Horwitz received her PhD in biochemistry from Brandeis University. She has had a continuing interest in natural products as a source of new drugs for the treatment of cancer. Her most seminal research contribution has been in the development of Taxol(®). Dr. Horwitz and her colleagues made the discovery that Taxol had a unique mechanism of action and suggested that it was a prototype for a new class of antitumor drugs. Although Taxol was an antimitotic agent blocking cells in the metaphase stage of the cell cycle, Dr. Horwitz recognized that Taxol was blocking mitosis in a way different from that of other known agents. Her group demonstrated that the binding site for Taxol was on the ß-tubulin subunit. The interaction of Taxol with the ß-tubulin subunit resulted in stabilized microtubules, essentially paralyzing the cytoskeleton, thereby preventing cell division. Dr. Horwitz served as president (2002-2003) of the American Association for Cancer Research (AACR). She is a member of the National Academy of Sciences, the Institute of Medicine, the American Academy of Arts and Sciences, and the American Philosophical Society. She has received numerous honors and awards, including the C. Chester Stock Award from Memorial Sloan Kettering Cancer Center, the Warren Alpert Foundation Prize from Harvard Medical School, the Bristol-Myers Squibb Award for Distinguished Achievement in Cancer Research, the American Cancer Society's Medal of Honor, and the AACR Award for Lifetime Achievement in Cancer Research. The following interview was conducted on January 23, 2014.


Asunto(s)
Investigación Biomédica/historia , Descubrimiento de Drogas/historia , Farmacología/historia , Alcanos/historia , Antineoplásicos Fitogénicos/historia , Carbamatos/historia , Selección de Profesión , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Lactonas/historia , Terapia Molecular Dirigida/historia , Paclitaxel/historia , Pironas/historia , Moduladores de Tubulina/historia
16.
Am J Physiol Cell Physiol ; 308(8): C631-41, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25608532

RESUMEN

The proton-coupled folate transporter (PCFT) mediates intestinal folate absorption and transport of folates across the choroid plexus. This study focuses on the role of Tyr residues in PCFT function. The substituted Cys-accessibility method identified four Tyr residues (Y291, Y362, Y315, and Y414) that are accessible to the extracellular compartment; three of these (Y291, Y362, and Y315) are located within or near the folate binding pocket. When the Tyr residues were replaced with Cys or Ala, these mutants showed similar (up to 6-fold) increases in influx Vmax and Kt/Ki for [(3)H]methotrexate and [(3)H]pemetrexed. When the Tyr residues were replaced with Phe, these changes were moderated or absent. When Y315A PCFT was used as representative of the mutants and [(3)H]pemetrexed as the transport substrate, this substitution did not increase the efflux rate constant. Furthermore, neither influx nor efflux mediated by Y315A PCFT was transstimulated by the presence of substrate in the opposite compartment; however, substantial bidirectional transstimulation of transport was mediated by wild-type PCFT. This resulted in a threefold greater efflux rate constant for cells that express wild-type PCFT than for cells that express Y315 PCFT under exchange conditions. These data suggest that these Tyr residues, possibly through their rigid side chains, secure the carrier in a high-affinity state for its folate substrates. However, this may be achieved at the expense of constraining the carrier's mobility, thereby decreasing the rate at which the protein oscillates between its conformational states. The Vmax generated by these Tyr mutants may be so rapid that further augmentation during transstimulation may not be possible.


Asunto(s)
Ácido Fólico/metabolismo , Absorción Intestinal/fisiología , Transportador de Folato Acoplado a Protón/metabolismo , Tirosina/química , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Sitios de Unión/genética , Transporte Biológico/genética , Línea Celular Tumoral , Glutamatos/metabolismo , Guanina/análogos & derivados , Guanina/metabolismo , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Absorción Intestinal/genética , Metotrexato/metabolismo , Modelos Moleculares , Pemetrexed , Estructura Terciaria de Proteína , Transportador de Folato Acoplado a Protón/genética
17.
Cancer Chemother Pharmacol ; 73(5): 1055-62, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24682532

RESUMEN

PURPOSE: To investigate the impact of 5-formyltetrahydrofolate on the activities of pralatrexate, as compared to methotrexate (MTX), in vitro. METHODS: Cells were exposed to (6S)5-formyltetrahydrofolate (5-formylTHF) for 24 h, before or after a 6-h exposure to antifolates following which the cellular accumulation and activities of the drugs were evaluated in HeLa cells. RESULTS: A 24-h delay between a 6-h exposure to antifolates and a subsequent 24-h exposure to 4 µM 5-formylTHF sustained the full activities of both antifolates. A 72-h interval was required between a single exposure of up to 4 µM 5-formylTHF and subsequent exposure to drugs to sustain activities of the antifolates. When cells were incubated with 4 µM 5-formylTHF for 24 h weekly, for 4 weeks, there was no significant increase in the IC50 for pralatrexate, but the MTX IC50 increased 2.5-fold as compared to cells growing continuously in 25 nM 5-formylTHF. This cyclical exposure to 5-formylTHF increased the cell folate pool by 16 %, had no significant effect on the intracellular pralatrexate level, but decreased intracellular MTX by 15 %. An extracellular concentration of MTX 50-fold higher than that of pralatrexate was required to achieve an intracellular level, and growth inhibition, comparable to that of pralatrexate. CONCLUSIONS: Cyclical exposures to 5-formylTHF at levels in excess of what is achieved in most clinical "rescue" regimens do not affect pralatrexate accumulation nor antitumor activity in HeLa cells, in contrast to MTX. An important element in preserving pralatrexate activity is achieving a sufficient interval between exposure to 5-formylTHF and the next dose of antifolate.


Asunto(s)
Aminopterina/análogos & derivados , Ligasas de Carbono-Nitrógeno/metabolismo , Antagonistas del Ácido Fólico/farmacología , Metotrexato/farmacología , Neoplasias/tratamiento farmacológico , Aminopterina/administración & dosificación , Aminopterina/farmacología , Antagonistas del Ácido Fólico/administración & dosificación , Células HeLa , Humanos , Metotrexato/administración & dosificación , Neoplasias/metabolismo , Células Tumorales Cultivadas
18.
Annu Rev Physiol ; 76: 251-74, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24512081

RESUMEN

The properties of intestinal folate absorption were documented decades ago. However, it was only recently that the proton-coupled folate transporter (PCFT) was identified and its critical role in folate transport across the apical brush-border membrane of the proximal small intestine established by the loss-of-function mutations identified in the PCFT gene in subjects with hereditary folate malabsorption and, more recently, by the Pcft-null mouse. This article reviews the current understanding of the properties of PCFT-mediated transport and how they differ from those of the reduced folate carrier. Other processes that contribute to the transport of folates across the enterocyte, along with the contribution of the enterohepatic circulation, are considered. Important unresolved issues are addressed, including the mechanism of intestinal folate absorption in the absence of PCFT and regulation of PCFT gene expression. The impact of a variety of ions, organic molecules, and drugs on PCFT-mediated folate transport is described.


Asunto(s)
Ácido Fólico/metabolismo , Absorción Intestinal/fisiología , Animales , Circulación Enterohepática/fisiología , Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/metabolismo , Humanos , Absorción Intestinal/genética , Intestino Grueso/metabolismo , Intestino Delgado/metabolismo , Síndromes de Malabsorción/genética , Síndromes de Malabsorción/metabolismo , Ratones , Transportador de Folato Acoplado a Protón/genética , Transportador de Folato Acoplado a Protón/metabolismo
19.
Mol Pharmacol ; 85(2): 310-21, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24249723

RESUMEN

The reduced folate carrier (RFC), proton-coupled folate transporter (PCFT), and folate receptors (FR) are folate-specific transporters. Antifolates currently in the clinic, such as pemetrexed, methotrexate, and pralatrexate, are transported into tumor cells primarily via RFC. Folic acid conjugated to cytotoxics, a new class of antineoplastics, are transported into cells via FR-mediated endocytosis. To better define the role of PCFT in antifolate resistance, a methotrexate-resistant cell line, M160-8, was selected from a HeLa subline in which the RFC gene was deleted and PCFT was highly overexpressed. These cells were cross-resistant to pemetrexed. PCFT function and the PCFT mRNA level in M160-8 cells were barely detectable, and FR-α function and mRNA level were increased as compared with the parent cells. While pemetrexed rapidly associated with FR and was internalized within endosomes in M160-8 cells, consistent with FR-mediated transport, subsequent pemetrexed and (6S)-5-formyltetrahydrofolate export into the cytosol was markedly impaired. In contrast, M160-8 cells were collaterally sensitive to EC0905, a folic acid-desacetylvinblastine monohydrazide conjugate also transported by FR-mediated endocytosis. However, in this case a sulfhydryl bond is cleaved to release the lipophilic cytotoxic moiety into the endosome, which passively diffuses out of the endosome into the cytosol. Hence, resistance to pemetrexed in M160-8 cells was due to entrapment of the drug within the endosome due to the absence of PCFT under conditions in which the FR cycling function was intact.


Asunto(s)
Antineoplásicos/farmacología , Endocitosis , Antagonistas del Ácido Fólico/farmacología , Transportadores de Ácido Fólico/fisiología , Ácido Fólico/farmacología , Glutamatos/farmacología , Guanina/análogos & derivados , Vinblastina/análogos & derivados , Células Cultivadas , Resistencia a Antineoplásicos , Transportadores de Ácido Fólico/análisis , Guanina/farmacología , Humanos , Pemetrexed , Transportador de Folato Acoplado a Protón/genética , Transportador de Folato Acoplado a Protón/fisiología , Vinblastina/farmacología
20.
Cancer Chemother Pharmacol ; 72(3): 597-606, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23881211

RESUMEN

PURPOSE: To characterize, directly and for the first time, the membrane transport and metabolism of pralatrexate, a new-generation dihydrofolate reductase inhibitor approved for the treatment for peripheral T-cell lymphoma. EXPERIMENTAL DESIGN: [(3)H]pralatrexate transport was studied in unique HeLa cell lines that express either the reduced folate carrier (RFC) or the proton-coupled folate transporter (PCFT). Metabolism to active polyglutamate derivatives was assessed by liquid chromatography. These properties were compared to those of methotrexate (MTX). RESULTS: The pralatrexate influx K t, mediated by RFC, the major route of folate/antifolate transport at systemic pH, was 0.52 µΜ, 1/10th the MTX influx K i. The electrochemical potential of pralatrexate within HeLa cells far exceeded the extracellular level and was greater than for MTX. In contrast, MTX transport mediated by PCFT, the mechanism of folate/antifolate absorption in the small intestine, exceeded that for pralatrexate. After a 6 h exposure of HeLa cells to 0.5 µM pralatrexate, 80 % of intracellular drug was its active polyglutamate forms, predominantly the tetraglutamate, and was suppressed when cells were loaded with natural folates. There was negligible formation of MTX polyglutamates. The difference in pralatrexate and MTX growth inhibition was far greater after transient exposures (375-fold) than continuous exposure (25-fold) to the drugs. CONCLUSIONS: Pralatrexate's enhanced activity relative to MTX is due to its much more rapid rate of transport and polyglutamation, the former less important when the carrier is saturated. The low affinity of pralatrexate for PCFT predicts a lower level of enterohepatic circulation and increased fecal excretion of the drug relative to MTX.


Asunto(s)
Aminopterina/análogos & derivados , Antagonistas del Ácido Fólico/farmacocinética , Metotrexato/farmacocinética , Transportador de Folato Acoplado a Protón/metabolismo , Proteína Portadora de Folato Reducido/metabolismo , Aminopterina/farmacocinética , Transporte Biológico , Cromatografía Liquida/métodos , Electroquímica , Células HeLa , Humanos , Ácido Poliglutámico/química , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...