Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Metabolism ; 110: 154306, 2020 09.
Article in English | MEDLINE | ID: mdl-32621820

ABSTRACT

BACKGROUND: Iron is finely regulated due to its vital roles in organisms and the peroxidase reactivity if excess. Solute Carrier Family 46 Member 1 (SLC46A1), also named PCFT or HCP1, is the main importer of heme­iron in the intestine, but has a high abundance in the liver. Since the liver has a central role in iron homeostasis, whether SLC46A1 regulates hepatic iron metabolism is of interest to be identified. METHODS: The recombinant adeno-associated virus vectors were used to hepatic-specifically inhibit SLC46A1 expression to observe its effects on hepatic iron metabolism. Then the abilities of SLC46A1 in importing heme and folate, and consequent alterations of iron content in hepatocytes were determined. Furthermore, effects of iron on SLC46A1 expression were investigated both in vitro and in vivo. RESULTS: The hepatocyte-specific inhibition of SLC46A1 decreases iron content in the liver and increases iron content in serum. Expressions of iron-related molecules, transferrin receptor 1, hepcidin and ferroportin, are correspondingly altered. Interestingly, free heme concentration in serum is increased, indicating a decreased import of heme by the liver. In hepatocytes, SLC46A1 is capable of importing hemin, increasing intracellular iron content. The import of hemin by SLC46A1 is unaffected by its other substrate, folate. Instead, hemin treatment decreases SLC46A1 expression, reducing the import of folate. In addition, SLC46A1 itself shows to be iron-responsive both in vivo and in vitro, making it available for regulating iron metabolism. CONCLUSION: The results elucidate that SLC46A1 regulates iron metabolism in the liver through a folate-independent manner of importing heme. The iron-responsive characters of SLC46A1 give us a new clue to link heme or iron overload with folate deficiency diseases.


Subject(s)
Heme/metabolism , Iron/metabolism , Liver/metabolism , Proton-Coupled Folate Transporter/physiology , Animals , Cells, Cultured , Hemin/metabolism , Hepatocytes/metabolism , Humans , Mice , Mice, Inbred C57BL , Proton-Coupled Folate Transporter/antagonists & inhibitors
2.
Cancer Chemother Pharmacol ; 81(1): 1-15, 2018 01.
Article in English | MEDLINE | ID: mdl-29127457

ABSTRACT

This review considers the "promise" of exploiting the proton-coupled folate transporter (PCFT) for selective therapeutic targeting of cancer. PCFT was discovered in 2006 and was identified as the principal folate transporter involved in the intestinal absorption of dietary folates. The recognition that PCFT was highly expressed in many tumors stimulated substantial interest in using PCFT for cytotoxic drug targeting, taking advantage of its high level transport activity under the acidic pH conditions that characterize many tumors. For pemetrexed, among the best PCFT substrates, transport by PCFT establishes its importance as a clinically important transporter in malignant pleural mesothelioma and non-small cell lung cancer. In recent years, the notion of PCFT-targeting has been extended to a new generation of tumor-targeted 6-substituted pyrrolo[2,3-d]pyrimidine compounds that are structurally and functionally distinct from pemetrexed, and that exhibit near exclusive transport by PCFT and potent inhibition of de novo purine nucleotide biosynthesis. Based on compelling preclinical evidence in a wide range of human tumor models, it is now time to advance the most optimized PCFT-targeted agents with the best balance of PCFT transport specificity and potent antitumor efficacy to the clinic to validate this novel paradigm of highly selective tumor targeting.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Molecular Targeted Therapy , Proton-Coupled Folate Transporter/antagonists & inhibitors , Acidosis , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Hypoxia , Folic Acid Antagonists/administration & dosage , Folic Acid Antagonists/therapeutic use , Humans , Lung Neoplasms/metabolism , Mesothelioma/metabolism , Mesothelioma, Malignant , Pleural Neoplasms/drug therapy , Pleural Neoplasms/metabolism , Proton-Coupled Folate Transporter/metabolism , Purine Nucleotides/biosynthesis , Reduced Folate Carrier Protein/metabolism
3.
Drug Metab Pharmacokinet ; 32(6): 311-314, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29162466

ABSTRACT

Myricetin is a flavonoid that inhibits human proton-coupled folate transporter (hPCFT) in a transient manner, in which inhibition is manifested in its presence, and also in a sustained manner, in which inhibition induced in its presence persists after its removal. In an effort to elucidate the mechanisms involved in those, we examined if myricetin might or might not act similarly on some other transporters. Transporters examined for that, in comparison with hPCFT, were its rat ortholog (rPCFT) and human riboflavin transporter 3 (hRFVT3). Experiments were conducted, using human embryonic kidney 293 cells transiently expressing the transporter to be examined, to assess the effects of myricetin (100 µM) on the uptake of folate by the PCFTs and riboflavin by hRFVT3. For hPCFT, myricetin was confirmed to induce a transient inhibition and also a sustained inhibition. However, myricetin induced neither transient nor sustained type of rPCFT inhibition. hRFVT3 was inhibited by myricetin in a transient manner, but not in a sustained manner. These results suggest the involvement of a hPCFT-specific mechanism in the sustained inhibition. The transient inhibition may be induced by a mechanism specific to hPCFT and also hRFVT3.


Subject(s)
Flavonoids/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Proton-Coupled Folate Transporter/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Flavonoids/metabolism , Folic Acid/pharmacokinetics , HEK293 Cells , Humans , Membrane Transport Proteins/metabolism , Nerve Tissue Proteins/metabolism , Proton-Coupled Folate Transporter/metabolism , Rats , Receptors, G-Protein-Coupled , Riboflavin/pharmacokinetics , Structure-Activity Relationship
4.
Drug Metab Pharmacokinet ; 30(5): 341-6, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26403086

ABSTRACT

Myricetin is a flavonoid that has recently been suggested to induce sustained inhibition of proton-coupled folate transporter (PCFT/SLC46A1), which operates for intestinal folate uptake. The present study was conducted to characterize the inhibitory effect in more detail, using human PCFT stably expressed in Madin-Darby canine kidney II cells, to gain information to cope with problems potentially arising from that. The kinetics of saturable folate transport was first assessed in the absence of myricetin in the cells pretreated with the flavonoid for 60 min. The pretreatment induced PCFT inhibition in a manner dependent on the concentration of myricetin, where the maximum transport rate was reduced by 35.5% and 83.1%, respectively, at its concentrations of 20 µM and 50 µM. The inhibitory effect was, however, less extensive at lower folate concentrations, because the Michaelis constant was also reduced similarly in a manner dependent on myricetin concentration. The inhibition was induced depending on the time of pretreatment and, after removal of myricetin (50 µM) upon the manifestation of an extensive inhibition at 60 min, reversed almost completely in 90 min. This rather short time required for recovery may suggest that the sustained inhibition of PCFT is of a reversible type.


Subject(s)
Flavonoids/pharmacology , Folic Acid Antagonists/pharmacology , Folic Acid/metabolism , Proton-Coupled Folate Transporter/antagonists & inhibitors , Androstadienes/pharmacology , Animals , Biological Transport, Active , Dogs , Dose-Response Relationship, Drug , Humans , Kinetics , Madin Darby Canine Kidney Cells , Protein Kinase Inhibitors/pharmacology , Wortmannin
5.
J Med Chem ; 58(17): 6938-59, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26317331

ABSTRACT

2-Amino-4-oxo-6-substituted-pyrrolo[2,3-d]pyrimidine antifolate thiophene regioisomers of AGF94 (4) with a thienoyl side chain and three-carbon bridge lengths [AGF150 (5) and AGF154 (7)] were synthesized as potential antitumor agents. These analogues inhibited proliferation of Chinese hamster ovary (CHO) sublines expressing folate receptors (FRs) α or ß (IC50s < 1 nM) or the proton-coupled folate transporter (PCFT) (IC50 < 7 nM). Compounds 5 and 7 inhibited KB, IGROV1, and SKOV3 human tumor cells at subnanomolar concentrations, reflecting both FRα and PCFT uptake. AGF152 (6) and AGF163 (8), 2,4-diamino-5-substituted-furo[2,3-d]pyrimidine thiophene regioisomers, also inhibited growth of FR-expressing CHO and KB cells. All four analogues inhibited glycinamide ribonucleotide formyltransferase (GARFTase). Crystal structures of human GARFTase complexed with 5 and 7 were reported. In severe combined immunodeficient mice bearing SKOV3 tumors, 7 was efficacious. The selectivity of these compounds for PCFT and for FRα and ß over the ubiquitously expressed reduced folate carrier is a paradigm for selective tumor targeting.


Subject(s)
Antineoplastic Agents/chemistry , Folate Receptor 1/antagonists & inhibitors , Folic Acid Antagonists/chemistry , Proton-Coupled Folate Transporter/antagonists & inhibitors , Pyrimidines/chemistry , Pyrroles/chemistry , Thiophenes/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Biological Transport , CHO Cells , Cell Line, Tumor , Cricetulus , Crystallography, X-Ray , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Female , Folate Receptor 1/chemistry , Folic Acid Antagonists/chemical synthesis , Folic Acid Antagonists/pharmacology , Heterografts , Humans , Mice, SCID , Models, Molecular , Neoplasm Transplantation , Pemetrexed/pharmacology , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Phosphoribosylglycinamide Formyltransferase/chemistry , Proton-Coupled Folate Transporter/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Pyrroles/chemical synthesis , Pyrroles/pharmacology , Stereoisomerism , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/pharmacology
6.
Biofactors ; 41(4): 232-41, 2015.
Article in English | MEDLINE | ID: mdl-26154406

ABSTRACT

Complex regulatory mechanisms control the expression of folate transporters within cells. Liver is the primary reserve of the folate stores within the body. As excessive alcohol consumption or inefficient dietary folate intake are known to create folate deficiency, so therefore the current study was designed to explore various regulatory mechanisms controlling the expression of folate transport in liver cells in conditions of ethanol exposure and folate deficiency. In order to see whether the effects mediated by the treatments are reversible or not, ethanol removal, and folate repletion was done after ethanol exposure and folate deficiency treatment respectively. Folate deficiency resulted an increase, whereas ethanol treatment decreased the folic acid uptake within the cells. The alterations in folic acid uptake were in agreement with the observed changes in the expression of folate transporters. Ethanol exposure resulted an increase in promoter methylation of reduced folate carrier; however, folate deficiency had no effect. The effects produced by ethanol exposure and folate deficiency were found to be reversible in nature as depicted in case of ethanol removal and folate repletion group. Rate of synthesis of folate transporters was found to be increased whereas half lives of mRNA of folate transporters was found to be decreased on folate deficiency treatment and reverse was the case on ethanol treatment. Overall, alteration in the expression of folate transporters under ethanol exposure and folate deficient conditions can be attributed to those regulatory mechanisms which work at the mRNA level.


Subject(s)
Ethanol/pharmacology , Folic Acid/pharmacology , Gene Expression Regulation/drug effects , Proton-Coupled Folate Transporter/genetics , RNA, Messenger/metabolism , Reduced Folate Carrier Protein/genetics , Biological Assay , Biological Transport , Cell Survival/drug effects , DNA Methylation/drug effects , Folate Receptor 1/agonists , Folate Receptor 1/antagonists & inhibitors , Folate Receptor 1/genetics , Folate Receptor 1/metabolism , Folic Acid Deficiency/genetics , Folic Acid Deficiency/metabolism , Folic Acid Deficiency/pathology , Hep G2 Cells , Humans , Lacticaseibacillus casei/growth & development , Lacticaseibacillus casei/metabolism , Models, Biological , Promoter Regions, Genetic , Proton-Coupled Folate Transporter/agonists , Proton-Coupled Folate Transporter/antagonists & inhibitors , Proton-Coupled Folate Transporter/metabolism , RNA, Messenger/agonists , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , Reduced Folate Carrier Protein/agonists , Reduced Folate Carrier Protein/antagonists & inhibitors , Reduced Folate Carrier Protein/metabolism , Signal Transduction , Tritium
7.
Drug Metab Pharmacokinet ; 30(2): 154-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25801697

ABSTRACT

Myricetin is a flavonoid that has recently been suggested to interfere with the intestinal folate transport system. To examine that possibility, focusing on its sustained inhibitory effect on proton-coupled folate transporter (PCFT), the uptake of folate was examined in Caco-2 cells, in which PCFT is known to be in operation, in the absence of myricetin in the medium during uptake period after preincubation of the cells with the flavonoid (100 µM) for 1 h. This pretreatment induced an extensive and sustained reduction in the carrier-mediated component of folate uptake, which was attributable to a reduction in the maximum transport rate (Vmax). Although the affinity of the transporter for folate was increased at the same time as indicated by a reduction in the Michaelis constant (Km), the change in Km was overwhelmed in extent by that in Vmax. Consistent with the finding, folate transport by human PCFT stably expressed in Madin-Darby canine kidney II cells was reduced in a similar manner with simultaneous reductions in Vmax and Km by myricetin pretreatment. Attention may need to be given for a possibility that such a sustained inhibition of PCFT could potentially be a cause of the malabsorption of folate and also antifolate drugs.


Subject(s)
Flavonoids/pharmacology , Folic Acid/metabolism , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Proton-Coupled Folate Transporter/antagonists & inhibitors , Animals , Biological Transport , Caco-2 Cells , Dogs , Dose-Response Relationship, Drug , Humans , Intestinal Mucosa/metabolism , Kinetics , Madin Darby Canine Kidney Cells , Models, Biological , Proton-Coupled Folate Transporter/genetics , Proton-Coupled Folate Transporter/metabolism , Transfection
8.
Drug Metab Pharmacokinet ; 29(4): 312-6, 2014.
Article in English | MEDLINE | ID: mdl-24492671

ABSTRACT

Myricetin is a flavonoid that has recently been suggested to interfere with the intestinal folate transport system. The present study was conducted to examine that possibility, focusing on its inhibitory effect on proton-coupled folate transporter (PCFT) as the molecular entity of the transport system. The uptake transport of folate was first examined in the Caco-2 cell as an intestinal epithelial cell model, and its carrier-mediated component, of which the Michaelis constant (Km) was 0.407 µM, was found to be noncompetitively inhibited by myricetin with an inhibition constant (Ki) of 61 µM. Consistent with that, folate transport by human PCFT stably expressed in Madin-Darby canine kidney II (MDCKII) cells, of which the Km was 1.246 µM, was also noncompetitively inhibited by myricetin with a Ki of 130 µM. Thus, myricetin was suggested to inhibit intestinal folate transport by acting noncompetitively on PCFT, although the Km and Ki were similarly shifted to some extent to be smaller in Caco-2 cells. Finally, epigallocatechin-3-gallate was also suggested to act in a noncompetitive manner as an inhibitory flavonoid. Care may need to be taken, therefore, in the ingestion of myricetin and some flavonoids to maintain the absorption of folate and antifolate drugs.


Subject(s)
Flavonoids/pharmacology , Folic Acid/metabolism , Proton-Coupled Folate Transporter/antagonists & inhibitors , Animals , Biological Transport/drug effects , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Line , Dogs , Folic Acid Antagonists/pharmacology , Humans , Methotrexate/metabolism
9.
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
10.
Pediatr Res ; 73(4 Pt 1): 388-94, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23338599

ABSTRACT

BACKGROUND: The mechanisms whereby gestational diabetes mellitus (GDM) increases the risk of fetal overgrowth and development of metabolic diseases later in life are likely to involve changes in nutrient supply to the fetus. Hence, in this work, we hypothesize that GDM may affect folic acid (FA) supply to the placenta and fetus. METHODS: We compared (3)H-FA uptake by human cytotrophoblasts isolated from normal pregnancies (normal trophoblasts; NTB cells) and GDM pregnancies (diabetic trophoblasts; DTB cells) and investigated the effect of GDM hallmarks on (3)H-FA uptake by BeWo cells. RESULTS: (3)H-FA uptake by NTB and DTB cells was time dependent and acidic pH stimulated. When compared with NTB, (3)H-FA uptake by DTB cells was more sensitive to acidic pH changes and to 5-methyltetrahydrofolate and pemetrexed (PTX) inhibition, indicating a proportionally greater involvement of the proton-coupled folate transporter (PCFT). A 4-h exposure of BeWo cells to lipopolysaccharide (LPS, 1-10 µg/ml) or to high levels of tumor necrosis factor-α (TNF-α, 300 ng/l) significantly reduced (3)H-FA uptake. Moreover, hyperleptinemic conditions (100 ng/ml leptin) decreased (3)H-FA uptake by BeWo cells in a time-dependent manner when compared with normoleptinemic conditions (1 ng/ml leptin). CONCLUSION: GDM modulates (3)H-FA uptake by the syncytiotrophoblast, and leptin as well as TNF-α downregulate it.


Subject(s)
Diabetes, Gestational/metabolism , Folic Acid/metabolism , Inflammation Mediators/metabolism , Leptin/metabolism , Trophoblasts/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adult , Biological Transport , Case-Control Studies , Cells, Cultured , Diabetes, Gestational/blood , Female , Glutamates/pharmacology , Guanine/analogs & derivatives , Guanine/pharmacology , Humans , Hydrogen-Ion Concentration , Kinetics , Lipopolysaccharides/pharmacology , Pemetrexed , Pregnancy , Proton-Coupled Folate Transporter/antagonists & inhibitors , Proton-Coupled Folate Transporter/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Tetrahydrofolates/pharmacology , Trophoblasts/drug effects
11.
Blood ; 117(18): 4895-904, 2011 May 05.
Article in English | MEDLINE | ID: mdl-21346251

ABSTRACT

The human proton coupled folate transporter (PCFT) is involved in low pH-dependent intestinal folate transport. In this report, we describe a new murine model of the hereditary folate malabsorption syndrome that we developed through targeted disruption of the first 3 coding exons of the murine homolog of the PCFT gene. By 4 weeks of age, PCFT-deficient (PCFT(-/-)) mice developed severe macrocytic normochromic anemia and pancytopenia. Immature erythroblasts accumulated in the bone marrow and spleen of PCFT(-/-) mice and failed to differentiate further, showing an increased rate of apoptosis in intermediate erythroblasts and reduced release of reticulocytes. In response to the inefficient hematologic development, the serum of the PCFT(-/-) animals contained elevated concentrations of erythropoietin, soluble transferrin receptor (sCD71), and thrombopoietin. In vivo folate uptake experiments demonstrated a systemic folate deficiency caused by disruption of PCFT-mediated intestinal folate uptake, thus confirming in vivo a critical and nonredundant role of the PCFT protein in intestinal folate transport and erythropoiesis. The PCFT-deficient mouse serves as a model for the hereditary folate malabsorption syndrome and is the most accurate animal model of folate deficiency anemia described to date that closely captures the spectrum of pathology typical of this disease.


Subject(s)
Folic Acid Deficiency/genetics , Folic Acid Deficiency/metabolism , Proton-Coupled Folate Transporter/antagonists & inhibitors , Proton-Coupled Folate Transporter/genetics , Animals , Base Sequence , Bone Marrow/pathology , DNA Primers/genetics , Disease Models, Animal , Erythropoiesis/genetics , Erythropoiesis/physiology , Female , Folic Acid/metabolism , Folic Acid Deficiency/pathology , Gene Targeting , Humans , Intestinal Absorption , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Pancytopenia/genetics , Pancytopenia/metabolism , Pancytopenia/pathology , Proton-Coupled Folate Transporter/deficiency , Spleen/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...