Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters

Publication year range
1.
Nature ; 595(7865): 130-134, 2021 07.
Article in English | MEDLINE | ID: mdl-34040256

ABSTRACT

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.


Subject(s)
Cryoelectron Microscopy , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/metabolism , Pemetrexed/chemistry , Pemetrexed/metabolism , Proton-Coupled Folate Transporter/chemistry , Proton-Coupled Folate Transporter/metabolism , Apoproteins/chemistry , Apoproteins/metabolism , Apoproteins/ultrastructure , Biological Transport , Humans , Models, Molecular , Proton-Coupled Folate Transporter/ultrastructure , Protons
2.
Proc Natl Acad Sci U S A ; 116(35): 17531-17540, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31405972

ABSTRACT

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.


Subject(s)
Brain/metabolism , Folate Receptor 1/metabolism , Folic Acid/metabolism , Reduced Folate Carrier Protein/metabolism , Vitamin D/metabolism , Animals , Biological Transport , Biomarkers , Blood-Brain Barrier/metabolism , Chromatography, Liquid , Female , Folate Receptor 1/genetics , Gene Expression , Immunohistochemistry , Mice , Mice, Knockout , Tandem Mass Spectrometry
3.
J Biol Chem ; 295(46): 15650-15661, 2020 11 13.
Article in English | MEDLINE | ID: mdl-32893190

ABSTRACT

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.


Subject(s)
Proton-Coupled Folate Transporter/metabolism , Amino Acid Sequence , Animals , Biological Transport , Cysteine/chemistry , Cysteine/metabolism , Folic Acid Deficiency/pathology , HeLa Cells , Humans , Kinetics , Malabsorption Syndromes/pathology , Methotrexate/metabolism , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Proton-Coupled Folate Transporter/chemistry , Proton-Coupled Folate Transporter/genetics
4.
J Biol Chem ; 294(18): 7245-7258, 2019 05 03.
Article in English | MEDLINE | ID: mdl-30858177

ABSTRACT

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)).


Subject(s)
Proton-Coupled Folate Transporter/metabolism , Cysteine/chemistry , Glycine/metabolism , HeLa Cells , Humans , Kinetics , Mutation , Protein Conformation , Proton-Coupled Folate Transporter/chemistry
5.
Am J Physiol Cell Physiol ; 314(3): C289-C296, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29167151

ABSTRACT

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.


Subject(s)
Folic Acid/metabolism , Proton-Coupled Folate Transporter/metabolism , Biological Transport , Cysteine , Disulfides/metabolism , HeLa Cells , Humans , Kinetics , Models, Molecular , Mutation , Oxidation-Reduction , Pemetrexed/metabolism , Protein Conformation, alpha-Helical , Protein Domains , Proton-Coupled Folate Transporter/chemistry , Proton-Coupled Folate Transporter/genetics , Structure-Activity Relationship
6.
Mol Pharmacol ; 93(3): 208-215, 2018 03.
Article in English | MEDLINE | ID: mdl-29326243

ABSTRACT

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.


Subject(s)
Folic Acid Antagonists/pharmacokinetics , Methotrexate/pharmacokinetics , Proton-Coupled Folate Transporter/metabolism , Biological Transport, Active , Buffers , HEPES/pharmacology , HeLa Cells , Humans , Hydrogen-Ion Concentration , Polyglutamic Acid/metabolism , Tumor Microenvironment
7.
Article in English | MEDLINE | ID: mdl-25562642

ABSTRACT

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.


Subject(s)
Biomedical Research/history , Drug Discovery/history , Pharmacology/history , Alkanes/history , Antineoplastic Agents, Phytogenic/history , Carbamates/history , Career Choice , History, 20th Century , History, 21st Century , Humans , Lactones/history , Molecular Targeted Therapy/history , Paclitaxel/history , Pyrones/history , Tubulin Modulators/history
8.
Annu Rev Physiol ; 76: 251-74, 2014.
Article in English | MEDLINE | ID: mdl-24512081

ABSTRACT

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.


Subject(s)
Folic Acid/metabolism , Intestinal Absorption/physiology , Animals , Enterohepatic Circulation/physiology , Folic Acid Deficiency/genetics , Folic Acid Deficiency/metabolism , Humans , Intestinal Absorption/genetics , Intestine, Large/metabolism , Intestine, Small/metabolism , Malabsorption Syndromes/genetics , Malabsorption Syndromes/metabolism , Mice , Proton-Coupled Folate Transporter/genetics , Proton-Coupled Folate Transporter/metabolism
9.
Am J Physiol Cell Physiol ; 312(4): C517-C526, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28122733

ABSTRACT

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.


Subject(s)
Amino Acid Substitution/physiology , Cysteine/chemistry , Cysteine/metabolism , Glutathione/chemistry , Glutathione/metabolism , Protein Processing, Post-Translational/physiology , Binding Sites , HeLa Cells , Humans , Protein Binding , Protein Engineering/methods , Structure-Activity Relationship
10.
J Biol Chem ; 291(15): 8162-72, 2016 Apr 08.
Article in English | MEDLINE | ID: mdl-26884338

ABSTRACT

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.


Subject(s)
Cysteine/chemistry , Disulfides/chemistry , Folic Acid/metabolism , Proton-Coupled Folate Transporter/chemistry , Proton-Coupled Folate Transporter/metabolism , Amino Acid Substitution , Cysteine/genetics , Cysteine/metabolism , Disulfides/metabolism , HeLa Cells , Humans , Models, Molecular , Mutation , Oxidation-Reduction , Protein Conformation , Protein Processing, Post-Translational , Protein Structure, Tertiary , Proton-Coupled Folate Transporter/genetics
11.
Biochim Biophys Acta Biomembr ; 1859(11): 2193-2202, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28802835

ABSTRACT

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.


Subject(s)
Cell Membrane/chemistry , Folic Acid/metabolism , Protein Interaction Domains and Motifs , Proton-Coupled Folate Transporter/chemistry , Proton-Coupled Folate Transporter/metabolism , Amino Acid Substitution , Binding Sites/genetics , Cell Membrane/metabolism , HeLa Cells , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Molecular , Mutant Proteins/chemistry , Protein Binding/genetics , Protein Domains/genetics , Protein Interaction Domains and Motifs/genetics , Protein Transport/genetics , Proton-Coupled Folate Transporter/genetics , Water/chemistry
12.
Mol Pharm ; 14(11): 3848-3858, 2017 11 06.
Article in English | MEDLINE | ID: mdl-28885847

ABSTRACT

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.


Subject(s)
Blood-Brain Barrier/metabolism , Receptors, Calcitriol/metabolism , Reduced Folate Carrier Protein/metabolism , Animals , Biological Transport , Brain/drug effects , Brain/metabolism , Calcitriol/pharmacology , Cells, Cultured , Folic Acid/metabolism , Humans , Male , Methotrexate/metabolism , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar , Receptors, Calcitriol/genetics , Reduced Folate Carrier Protein/genetics
13.
Am J Physiol Cell Physiol ; 311(1): C150-7, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27251438

ABSTRACT

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.


Subject(s)
Cell Membrane/metabolism , Folic Acid/metabolism , Proton-Coupled Folate Transporter/metabolism , Binding Sites , Biotinylation , Cell Membrane/drug effects , Cysteine , Folic Acid Antagonists/pharmacology , Genotype , HeLa Cells , Humans , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Kinetics , Mutagenesis, Site-Directed , Mutation , Pemetrexed/pharmacology , Phenotype , Protein Binding , Protein Conformation, alpha-Helical , Protein Domains , Protein Stability , Proton-Coupled Folate Transporter/chemistry , Proton-Coupled Folate Transporter/drug effects , Proton-Coupled Folate Transporter/genetics , Structure-Activity Relationship , Transfection , Tryptophan
14.
Am J Physiol Cell Physiol ; 308(8): C631-41, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25608532

ABSTRACT

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.


Subject(s)
Folic Acid/metabolism , Intestinal Absorption/physiology , Proton-Coupled Folate Transporter/metabolism , Tyrosine/chemistry , Amino Acid Sequence , Amino Acid Substitution , Binding Sites/genetics , Biological Transport/genetics , Cell Line, Tumor , Glutamates/metabolism , Guanine/analogs & derivatives , Guanine/metabolism , HeLa Cells , Humans , Hydrogen-Ion Concentration , Intestinal Absorption/genetics , Methotrexate/metabolism , Models, Molecular , Pemetrexed , Protein Structure, Tertiary , Proton-Coupled Folate Transporter/genetics
15.
Mol Pharmacol ; 85(2): 310-21, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24249723

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Endocytosis , Folic Acid Antagonists/pharmacology , Folic Acid Transporters/physiology , Folic Acid/pharmacology , Glutamates/pharmacology , Guanine/analogs & derivatives , Vinblastine/analogs & derivatives , Cells, Cultured , Drug Resistance, Neoplasm , Folic Acid Transporters/analysis , Guanine/pharmacology , Humans , Pemetrexed , Proton-Coupled Folate Transporter/genetics , Proton-Coupled Folate Transporter/physiology , Vinblastine/pharmacology
16.
Am J Physiol Cell Physiol ; 304(12): C1159-67, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23552283

ABSTRACT

The proton-coupled folate transporter (PCFT, SLC46A1) mediates folate transport across the apical brush-border membrane of the proximal small intestine and the basolateral membrane of choroid plexus ependymal cells. Two loss-of-function mutations in PCFT, which are the basis for hereditary folate malabsorption, have been identified within the fourth transmembrane domain (TMD4) in subjects with this disorder. We have employed the substituted Cys accessibility method (SCAM) to study the accessibilities of all residues in TMD4 and their roles in folate substrate binding to the carrier. When residues 146-167 were replaced by Cys, all except R148C were expressed at the cell surface. Modification of five of these substituted Cys residues (positions 147, 152, 157, 158, and 161) by methanethiosulfonate (MTS) reagents led to reduction of PCFT function. All five residues could be labeled with N-biotinylaminoethyl-MTS, and this could be blocked by the high-affinity PCFT substrate pemetrexed. Pemetrexed also protected PCFT mutant function from inhibitory modification of the substituted Cys at positions 157, 158, and 161 by a MTS. The findings indicate that these five residues in TMD4 are accessible to the aqueous translocation pathway, play a role in folate substrate binding, and are likely located within or near the folate binding pocket. A homology model of PCFT places three of these residues, Phe¹57, Gly¹58, and Leu¹6¹, within a breakpoint in the midportion of TMD4, a region that likely participates in alterations in the PCFT conformational state during carrier cycling.


Subject(s)
Amino Acid Substitution/genetics , Cysteine/chemistry , Cysteine/genetics , Proton-Coupled Folate Transporter/chemistry , Proton-Coupled Folate Transporter/genetics , Amino Acid Sequence , Gene Deletion , HeLa Cells , Humans , Molecular Sequence Data , Protein Structure, Tertiary/genetics , Protein Transport/genetics
17.
Mol Pharmacol ; 84(1): 95-103, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23609145

ABSTRACT

The proton-coupled folate transporter (PCFT) plays a key role in intestinal folate absorption, and loss-of-function mutations in the gene encoding this transporter are the molecular basis for hereditary folate malabsorption. Using a stable transfectant with high expression of PCFT, physiologic levels of bicarbonate produced potent and rapidly reversible inhibition of PCFT-mediated transport at neutral pH. Bisulfite and nitrite also inhibited PCFT function at neutral pH, whereas sulfate, nitrate, and phosphate had no impact at all. At weakly acidic pH (6.5), bisulfite and nitrite exhibited much stronger inhibition of PCFT-mediated transport, whereas sulfate and nitrate remained noninhibitory. Inhibition by bisulfite and nitrite at pH 6.5 was associated with a marked decrease in the influx Vmax and collapse of the transmembrane proton gradient attributed to the diffusion of the protonated forms into these cells. Monocarboxylates such as pyruvate and acetate also collapsed the pH gradient and were also inhibitory, whereas citrate and glycine neither altered the proton gradient nor inhibited PCFT-mediated transport. These observations add another dimension to the unfavorable pH environment for PCFT function in systemic tissues: the presence of high concentrations of bicarbonate.


Subject(s)
Bicarbonates/pharmacology , Proton-Coupled Folate Transporter/antagonists & inhibitors , Acetic Acid/pharmacology , Anions/pharmacology , Cell Line, Tumor , Citric Acid/pharmacology , Folic Acid Transporters/metabolism , Glycine/pharmacology , HeLa Cells , Humans , Hydrogen-Ion Concentration , Kinetics , Membrane Transport Proteins/metabolism , Nitrates/pharmacology , Proton-Coupled Folate Transporter/metabolism , Protons , Pyruvic Acid/pharmacology , Sulfites/pharmacology
18.
Am J Physiol Cell Physiol ; 303(6): C673-81, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22785121

ABSTRACT

The proton-coupled folate transporter (PCFT) mediates intestinal folate absorption, and loss-of-function mutations in this gene result in the autosomal recessive disorder hereditary folate malabsorption. The current study, focused on a structure-functional analysis of this transporter, identified Gly-189 and Gly-192 (a GxxG motif) located in the fifth transmembrane domain as residues that could not be replaced with alanine without a loss of function. In contrast, function was preserved when Gly-56 and Gly-59 (the other conservative GXXG motif in human PCFT) were replaced with alanine. Similarly, Gly-93 and Gly-97, which constitute the only conserved GXXXG dimerization motif in human PCFT, tolerated alanine substitution. To explore the role of this region in folate binding, the residues around Gly-189 and Gly-192 were analyzed by the substituted cysteine accessibility method. Both I188C and M193C mutants were functional and were inhibited by membrane-impermeable sulfhydryl-reactive reagents; this could be prevented with PCFT substrate, but the protection was sustained at 0°C only for the I188C mutant, consistent with localization of Ile-188 in the PCFT folate binding pocket. The functional role of residues around Gly-189 and Gly-192 is consistent with a molecular structural model in which these two residues along with Ieu-188 are accessible to the PCFT aqueous translocation pathway.


Subject(s)
Folic Acid/metabolism , Glycine/physiology , Proton-Coupled Folate Transporter/chemistry , Proton-Coupled Folate Transporter/physiology , Amino Acid Motifs/physiology , Amino Acid Sequence , Binding Sites/physiology , Conserved Sequence , Folic Acid/chemistry , HeLa Cells , Humans , Isoleucine/genetics , Molecular Sequence Data , Protein Transport/physiology , Protons , Signal Transduction/physiology
19.
Am J Physiol Cell Physiol ; 302(9): C1405-12, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22345511

ABSTRACT

Proton-coupled folate transporter (PCFT) mediates folate intestinal absorption and transport across the choroid plexus, processes defective in subjects with hereditary folate malabsorption (HFM). PCFT is also widely expressed in human solid tumors where it contributes to the transport of pemetrexed and other antifolates. This study defines the basis for the functional changes due to a P425R mutation detected in a subject with HFM. Among various substitutions, only positively charged mutants (P425R and P425K) lost function but in a highly selective manner. Transport of reduced folates mediated by P425R-PCFT was virtually abolished; the methotrexate influx K(t) was increased fivefold (from 2 to 10 µM). In contrast, the pemetrexed influx K(t) mediated by P425R-PCFT was decreased 30% compared with wild-type (WT)-PCFT. Methotrexate inhibition of pemetrexed influx was competitive with a K(i) for WT-PCFT comparable to its influx K(t). However, the methotrexate influx K(i) for P425R-PCFT was ∼15-fold higher than the WT-PCFT influx K(t) and threefold higher than the methotrexate influx K(t) for the P425R-PCFT mutant. The confirmed secondary structure and homology modeling place the P425 residue at the junction of the 6th external loop and 12th transmembrane domain, remote from the aqueous translocation pathway, a prediction confirmed by the failure to label P425C-PCFT with N-biotinylaminoethyl methanethiosulfonate-biotin and the absence of inhibition of P425C-PCFT function by water-soluble sulfhydryl reagents. Hence, despite its location, the P425R-PCFT mutation produces a conformational change that fully preserves pemetrexed binding but markedly impairs binding of methotrexate and other folates to the carrier.


Subject(s)
Folic Acid/metabolism , Malabsorption Syndromes/genetics , Mutation , Proton-Coupled Folate Transporter/genetics , Proton-Coupled Folate Transporter/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Blotting, Western , Cells, Cultured , Glutamates/chemistry , Glutamates/pharmacokinetics , Guanine/analogs & derivatives , Guanine/chemistry , Guanine/pharmacokinetics , Humans , Immunoprecipitation , Malabsorption Syndromes/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Pemetrexed , Protein Binding , Protein Structure, Secondary , Proton-Coupled Folate Transporter/chemistry , Transfection
20.
Mol Pharmacol ; 82(2): 209-16, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22554803

ABSTRACT

5-Aminoimidazole-4-carboxamide riboside (AICAR), an agent with diverse pharmacological properties, augments transport of folates and antifolates. This report further characterizes this phenomenon and defines the mechanism by which it occurs. Exposure of HeLa cells to AICAR resulted in augmentation of methotrexate, 5-formyltetrahydrofolate, and 5-methyltetrahydrofolate initial rates and net uptake in cells that express the reduced folate carrier (RFC). This did not occur in cells that express only the proton-coupled folate transporter and accumulated folates by this mechanism. Transport stimulation correlated with the accumulation of 5-aminoimidazole-4-carboxamide ribotide monophosphate (ZMP), the monophosphate derivative of AICAR, within cells as established by liquid chromatography. When ZMP formation was blocked with 5-iodotubercidin, an inhibitor of adenosine kinase, folate transport stimulation by AICAR was absent. When cells first accumulated ZMP and were then exposed to 5-iodotubercidin or AICAR-free buffer, the ZMP level markedly decreased and folate transport stimulation was abolished. Extracellular ZMP inhibited RFC-mediated folate influx, and the presence of intracellular ZMP correlated with inhibition of folate efflux. The data indicate that intracellular ZMP trans-stimulates folate influx and inhibits folate efflux, which, together, produce a marked augmentation in the net cellular folate level. This interaction among ZMP, folates, and RFC, a folate/organic phosphate antiporter, is consistent with a classic exchange reaction. The transmembrane gradient for one transport substrate (ZMP) drives the uphill transport of another (folate) via a carrier used by both substrates, a phenomenon intrinsic to the energetics of RFC-mediated folate transport.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Folic Acid Antagonists/agonists , Folic Acid Antagonists/metabolism , Reduced Folate Carrier Protein/agonists , Reduced Folate Carrier Protein/metabolism , Ribonucleotides/metabolism , Ribonucleotides/pharmacology , Aminoimidazole Carboxamide/metabolism , Aminoimidazole Carboxamide/pharmacology , Drug Synergism , HeLa Cells , Humans , Protein Transport/drug effects , Protein Transport/physiology
SELECTION OF CITATIONS
SEARCH DETAIL