Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 380(1): 5-10, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19138668

ABSTRACT

We have previously shown that overexpression of the multidrug resistance (MDR) efflux transporter ABCG2 in the membrane of novel extracellular vesicles that are confined to breast cancer cell-cell attachment zones confers mitoxantrone resistance and mediates a marked intravesicular concentration of an unknown endogenous green fluorescent compound (I. Ifergan, G.L. Scheffer, Y.G. Assaraf, Novel extracellular vesicles mediate an ABCG2-dependent anticancer drug sequestration and resistance, Cancer Res. 65 (2005) 10952-10958). Here we identified the latter as riboflavin (vitamin B2) and further demonstrated that the marked intravesicular concentration of riboflavin in ABCG2-overexpressing breast and lung cancer cells tightly correlates with the extent of ABCG2 overexpression and its differential localization to the vesicular membrane and not to the plasma membrane surrounded by growth medium. We hence propose that the ABCG2-dependent concentration of riboflavin in these intercellular compartments may serve as a novel, sensitive, and non-cytotoxic (i.e. based on vitamin accumulation) functional marker for the quantification of the levels of MDR mediated by ABCG2-rich extracellular vesicles in multiple malignant cells.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Neoplasm Proteins/biosynthesis , Riboflavin/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Breast Neoplasms/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Female , Humans , Lung Neoplasms/metabolism
2.
Vitam Horm ; 79: 99-143, 2008.
Article in English | MEDLINE | ID: mdl-18804693

ABSTRACT

Folic acid is an essential vitamin for a wide spectrum of biochemical reactions; however, unlike bacteria and plants, mammals are devoid of folate biosynthesis and thus must obtain this cofactor from exogenous sources. Therefore, folate deficiency may impair the de novo biosynthesis of purines and thymidylate and thereby disrupt DNA and RNA metabolism, homocysteine remethylation, methionine biosynthesis, and subsequent formation of S-adenosylmethionine (the universal methyl donor) which in turn may lead to altered methylation reactions. This impaired folate-dependent intracellular metabolism can lead to several key pathologies including, for example, megaloblastic anemia, homocysteinemia, cardiovascular disease, embryonic defects, in particular neural tube defects (NTDs), congenital heart defects, and possibly cancer. The current review presents and evaluates the up-to-date knowledge regarding the molecular mechanisms underlying cellular survival and/or adaptation to folate deficiency or insufficiency. These mechanisms of adaptation to folate deficiency generally associated with folate uptake, intracellular folate retention, folate-dependent metabolism, and active folate efflux specifically include: (a) Up- or downregulation of various folate-dependent enzymes like dihydrofolate reductase (DHFR) and thymidylate synthase (TS), (b) Cellular retention of folates via polyglutamylation by the enzyme folylpoly-gamma-glutamate synthetase (FPGS), (c) Overexpression of folate influx systems including the reduced folate carrier (RFC), folate receptor (FR) as well as the proton-coupled folate transporter (PCFT), a recently identified intestinal folate influx transporter optimally functioning at the acidic microclimate of the upper intestinal epithelium, (d) Downregulation of ATP-driven folate efflux transporters of the multidrug resistance protein (MRP; ABCC) family and breast cancer resistance protein (BCRP; ABCG2) that belong to the multidrug resistance (MDR) efflux transporters of the ATP-binding cassette (ABC) superfamily. Moreover, the intricate interplay between various components of the adaptive response to folate deprivation is also discussed.


Subject(s)
Folic Acid Deficiency/metabolism , Folic Acid/metabolism , Animals , Antineoplastic Agents/pharmacology , Atherosclerosis , Carrier Proteins/genetics , Carrier Proteins/metabolism , Drug Resistance, Neoplasm , Gene Expression Regulation , Heart Defects, Congenital , Humans , Hyperhomocysteinemia , Mice , Neoplasms/drug therapy , Neural Tube Defects
3.
J Biol Chem ; 283(30): 20687-95, 2008 Jul 25.
Article in English | MEDLINE | ID: mdl-18499665

ABSTRACT

The reduced folate carrier (RFC), a bidirectional anion transporter, is the major uptake route of reduced folates essential for a spectrum of biochemical reactions and thus cellular proliferation. However, here we show that ectopic overexpression of the RFC, but not of folate receptor alpha, a high affinity unidirectional folate uptake route serving here as a negative control, resulted in an approximately 15-fold decline in cellular viability in medium lacking folates but not in folate-containing medium. Moreover to explore possible mechanisms of adaptation to folate deficiency in various cell lines that express the endogenous RFC, we first determined the gene expression status of the following genes: (a) RFC, (b) ATP-driven folate exporters (i.e. MRP1, MRP5, and breast cancer resistance protein), and (c) folylpoly-gamma-glutamate synthetase and gamma-glutamate hydrolase (GGH), enzymes catalyzing folate polyglutamylation and hydrolysis, respectively. Upon 3-7 days of folate deprivation, semiquantitative reverse transcription-PCR analysis revealed a specific approximately 2.5-fold decrease in RFC mRNA levels in both breast cancer and T-cell leukemia cell lines that was accompanied by a consistent fall in methotrexate influx, serving here as an RFC transport activity assay. Likewise a 2.4-fold decrease in GGH mRNA levels and approximately 19% decreased GGH activity was documented for folate-deprived breast cancer cells. These results along with those of a novel mathematical biomodeling devised here suggest that upon severe short term (i.e. up to 7 days) folate deprivation RFC transport activity becomes detrimental as RFC, but not ATP-driven folate exporters, efficiently extrudes folate monoglutamates out of cells. Hence down-regulation of RFC and GGH may serve as a novel adaptive response to severe folate deficiency.


Subject(s)
Folic Acid/chemistry , Gene Expression Regulation, Neoplastic , Membrane Transport Proteins/physiology , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Survival , Cricetinae , Cricetulus , Humans , Kinetics , Membrane Transport Proteins/metabolism , Models, Biological , Models, Theoretical , Oligonucleotides/chemistry , Reduced Folate Carrier Protein , Reverse Transcriptase Polymerase Chain Reaction
4.
Biochem Pharmacol ; 74(1): 41-53, 2007 Jun 30.
Article in English | MEDLINE | ID: mdl-17481587

ABSTRACT

The A421 ABCG2 genotype is a frequent polymorphism encoding the K141 transporter, which is associated with a significant decrease in transporter expression and function when compared to the wild type (wt) C421 allele encoding the Q141 ABCG2. Here we show that during the acquisition of resistance to the novel triazoloacridone antitumor agent C-1305 in lung cancer cells harboring a heterozygous C421A genotype, a marked C421 allele-specific ABCG2 gene amplification occurred. This monoallelic C421 ABCG2 gene amplification brought about the overexpression of both C421 ABCG2 mRNA and the transporter at the plasma membrane. This resulted in the lack of cellular drug accumulation due to increased efflux of both C1305 and C-1311, a fluorescent imidazoacridone homologue of C-1305, as well as marked resistance to these antitumor agents and to established ABCG2 substrates including mitoxantrone and SN-38. Consistently, the accumulation and sensitivity to these drugs were restored upon incubation with the potent and specific ABCG2 transport inhibitors Ko143 and fumitremorgin C. Moreover, upon transfection into HEK293 cells, the wt Q141 ABCG2 allele displayed a significantly decreased accumulation of C-1311 and increased resistance to C-1305, C-1311 and mitoxantrone, when compared to the K141 ABCG2 transfectant. Hence, the current study provides the first evidence that during the exposure to anticancer drugs, an allele-specific Q141 ABCG2 gene amplification occurs that confers a drug resistance advantage when compared to the K141 ABCG2. These findings have important implications for the selection and expansion of malignant anticancer drug resistant clones during chemotherapy with ABCG2 drugs.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Acridines/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Drug Resistance, Neoplasm/genetics , Gene Amplification , Neoplasm Proteins/genetics , Triazoles/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/metabolism , Acridines/metabolism , Aminoacridines/metabolism , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Multiple/drug effects , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic , Heterozygote , Humans , Indoles/pharmacology , Irinotecan , Mitoxantrone/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , RNA, Messenger/metabolism , Triazoles/metabolism
5.
Ann Rheum Dis ; 66(10): 1289-95, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17267514

ABSTRACT

BACKGROUND: Glucocorticoids (GCs) are commonly used in the treatment of (chronic) inflammatory diseases and cancer, but inherent or acquired resistance to these drugs limits their optimal efficacy. The availability of drugs that could modulate GC resistance is therefore of potential clinical interest. OBJECTIVE: To explore the molecular basis of GC sensitisation of GC resistant monocytic/macrophage cells after chronic exposure to sulfasalazine. METHODS: Human monocytic/macrophage THP1 and U937 cells represent a cell line model system characterised by inherent resistance to the GCs dexamethasone and prednisolone. Both cell lines were chronically exposed in vitro to 0.3-0.6 mM sulfasalazine (SSZ) for approximately 3 months, after which they were characterised for GC sensitivity, expression levels of GC receptor and components of the nuclear factor kappa B (NFkappaB) signalling pathway, and their ability to undergo GC induced apoptosis. RESULTS: Chronic exposure to SSZ markedly sensitised both U937 and THP1 cells to dexamethasone (781-fold and 1389-fold, respectively) and prednisolone (562-fold and 1220-fold, respectively). Restoration of GC sensitivity in cells exposed to SSZ was provoked via GC induced apoptosis, coinciding with inhibition of NFkappaB activation. Moreover, western blot analysis revealed a markedly increased expression of glucocorticoid receptor alpha (GRalpha) in cells exposed to SSZ. Since GRalpha mRNA levels were only marginally increased, these results suggest that an altered post-transcriptional mechanism was operable which conferred a stable GRalpha protein on SSZ exposed cells. CONCLUSION: These results suggest that chronic targeting of the NFkappaB signalling pathway by SSZ may be exploited as a novel strategy to stabilise GRalpha expression and thereby sensitise primary resistant cells to GCs.


Subject(s)
Apoptosis/drug effects , Glucocorticoids/pharmacology , Macrophages/drug effects , Monocytes/drug effects , Receptors, Glucocorticoid/metabolism , Sulfasalazine/pharmacology , Cell Line , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Drug Resistance , Humans , NF-kappa B/antagonists & inhibitors , Prednisolone/pharmacology , Signal Transduction/physiology , U937 Cells , Up-Regulation/physiology
6.
Cancer Chemother Pharmacol ; 58(6): 826-34, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16612649

ABSTRACT

Cellular uptake of hydrophilic antifolates proceeds via the reduced folate carrier whereas lipophilic antifolates enter cells by diffusion. Recently we have shown that transfectant cells overexpressing the mutant G482 ABCG2 displayed 120-6,250-fold resistance to hydrophilic antifolates than untransfected cells upon 4 h drug exposure, but lost almost all their antifolate resistance upon 72 h drug exposure (Shafran et al. in Cancer Res 65:8414-8422, 2005). Here we explored the ability of the wild type (WT) R482-as well as the mutant G482-and T482 ABCG2 to confer resistance to lipophilic antifolate inhibitors of dihydrofolate reductase (trimetrexate, piritrexim, metoprine and pyrimethamine) and thymidylate synthase (AG337, AG377 and AG331). Lipophilic antifolate resistance was determined using growth inhibition assays upon 72 h drug exposure. Cells overexpressing these mutant efflux transporters displayed up to 106-fold resistance to lipophilic antifolates relative to untransfected cells; this resistance was reversed by the specific and potent ABCG2 efflux inhibitor Ko143. In contrast, cells overexpressing the WT R482 ABCG2 exhibited either no or only a low-level of lipophilic antifolate resistance. These results provide the first evidence that overexpression of the mutant G482- and T482 but not the WT R482 ABCG2 confers a high-level of resistance to lipophilic antifolates. The high membrane partitioning of lipophilic antifolates along with the large confinement of ABCG2 to the plasma membrane suggest that these mutant ABCG2 transporters may possibly recognize and extrude lipophilic antifolates from the lipid bilayer. The potential implications to cancer chemotherapy as well as the mechanism of anticancer drug extrusion by these mutant exporters are discussed.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Drug Resistance, Neoplasm/genetics , Folic Acid Antagonists/pharmacology , Mutation/genetics , Neoplasm Proteins/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biological Transport/drug effects , Biological Transport/genetics , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cisplatin/chemistry , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Doxorubicin/chemistry , Doxorubicin/pharmacology , Fluorouracil/chemistry , Fluorouracil/pharmacology , Folic Acid Antagonists/chemistry , Heterocyclic Compounds, 3-Ring/metabolism , Humans , Indoles/chemistry , Indoles/pharmacology , Lipids/chemistry , Molecular Structure , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Paclitaxel/chemistry , Paclitaxel/pharmacology , Pyrimethamine/analogs & derivatives , Pyrimethamine/chemistry , Pyrimethamine/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Quinazolines/chemistry , Quinazolines/pharmacology , Rhodamines , Transfection , Trimetrexate/chemistry , Trimetrexate/pharmacology
7.
Arthritis Rheum ; 54(2): 557-68, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16447232

ABSTRACT

OBJECTIVE: To explore the onset and molecular mechanism of resistance to the antimalarial disease-modifying antirheumatic drug (DMARD) chloroquine (CQ) in human CEM T cells. METHODS: Human CEM cells were used as an in vitro model system to study the development of CQ resistance by growing cells in stepwise increasing concentrations of CQ. RESULTS: Over a period of 6 months, CEM cell lines developed 4-5-fold resistance to CQ. CQ resistance was associated with the specific overexpression of multidrug resistance-associated protein 1 (MRP-1), an ATP-driven drug efflux pump. This was illustrated by 1) overexpression of MRP-1 by Western blotting and 2) the complete reversal of CQ resistance by the MRP-1 transport inhibitors MK571 and probenecid. Importantly, CQ-resistant CEM cells retained full sensitivity to other DMARDs, including methotrexate, leflunomide, cyclosporin A, and sulfasalazine, but exhibited a high level of cross-resistance (>1,000-fold) to the glucocorticoid dexamethasone. The mechanistic basis for the latter was associated with aberrant signaling via the cAMP-protein kinase A pathway, since the cAMP-inducing agent forskolin reversed dexamethasone resistance. Finally, CQ-resistant CEM cells displayed a markedly reduced capacity to release proinflammatory cytokines (tumor necrosis factor alpha) and chemokines (interleukin-8). CONCLUSION: Induction of overexpression of the multidrug resistance efflux transporter MRP-1 can emerge after long-term exposure to CQ and results in CQ resistance and collateral resistance to dexamethasone. These findings warrant further detailed investigations into the possible role of MRP-1 and other members of the superfamily of drug efflux pumps in diminishing the efficacy of DMARDs in rheumatoid arthritis treatment.


Subject(s)
Antirheumatic Agents/pharmacology , Chloroquine/pharmacology , Drug Resistance, Multiple/drug effects , Glucocorticoids/pharmacology , Multidrug Resistance-Associated Proteins/metabolism , T-Lymphocytes/drug effects , Blotting, Western , Cell Line , Dose-Response Relationship, Drug , Humans , T-Lymphocytes/metabolism
8.
J Theor Biol ; 240(4): 637-47, 2006 Jun 21.
Article in English | MEDLINE | ID: mdl-16352313

ABSTRACT

Antifolates are used in the treatment of various human malignancies and exert their cytotoxic activity by inhibiting folate-dependent enzymes resulting in disruption of DNA synthesis and cell death. Here we devised a computerized hybrid functional petri nets (HFPN) modelling of folate metabolism under physiological and antifolate inhibitory conditions. This HFPN modelling proved valid as a good agreement was found between the simulated steady-state concentrations of various reduced folates and those published for cell extracts; consistently, the simulation derived total folate pool size (11.3 microM) was identical to that published for cell extracts. In silico experiments were conducted to characterize the inhibitory profile of four distinct antifolates including methotrexate (MTX), tomudex, and LY309887, which inhibit dihydrofolate reductase (DHFR), thymidylate synthase (TS) and glycineamide ribonucleotide transformylase (GARTFase), respectively, as well as pemetrexed which has the capacity to inhibit all three enzymes. In order to assess the inhibitory activity of antifolates on purines and pyrimidines, the biosynthesis rates of IMP (20.53 microM/min) and dTMP (23.8 microM/min) were first simulated. Whereas the biochemical inhibitory profile of MTX was characterized by increased dihydrofolate and decreased tetrahydrofolate (THF) concentrations, the remaining antifolates did not decrease THF levels. Furthermore, MTX was 766- and 10-fold more potent in decreasing the production rates of IMP and dTMP, respectively, than pemetrexed. LY309887 indirectly decreased the rate of dTMP production by reducing the levels of 5-CH2-THF, a folate cofactor for TS. Surprisingly, pemetrexed failed to inhibit DHFR even at high concentrations. This HFPN-based simulation offers an inexpensive, user-friendly, rapid and reliable means of pre-clinical evaluation of the inhibitory profiles of antifolates.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Computer Simulation , Folic Acid Antagonists/pharmacology , Folic Acid/metabolism , Models, Chemical , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Methotrexate/pharmacology , Purines/biosynthesis , Quinazolines/pharmacology , Signal Transduction , Tetrahydrofolates/pharmacology , Thiophenes/pharmacology , Thymidine Monophosphate/biosynthesis
9.
Blood ; 107(8): 3288-94, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16368880

ABSTRACT

The reduced folate carrier (RFC) is the dominant route for the uptake of various antifolates including PT523, a potent dihydrofolate reductase inhibitor (Ki = 0.35 pM) and an excellent transport substrate of the RFC (Kt = 0.7 microM). Here, we describe the multiple mechanisms of RFC inactivation in human leukemia PT523-resistant cells originally harboring 3 RFC alleles. Cellular exposure to gradually increasing PT523 concentrations resulted in sublines displaying up to 3500-fold resistance to various hydrophilic antifolates that rely on RFC for their cellular uptake. Antifolate-resistant cells lost RFC gene expression (65%-99% loss) due to impaired promoter binding of various transcription factors that regulate RFC gene expression. Additionally, DNA sequencing revealed that PT523-resistant cells contained a cluster of 4 nearly consecutive mutations residing on a single RFC allele including L143P, A147V, R148G, and Q150Stop. Southern blot analysis established the loss of an RFC allele in PT523-resistant cells. These alterations resulted in markedly decreased RFC protein levels (approximately 80%-99% loss) and consequently impaired [3H]methotrexate transport (87%-99% loss). This study provides the first evidence that acquisition of PT523 resistance in human leukemia cells harboring 3 RFC alleles is due to multiple coexisting alterations including transcriptional silencing, inactivating mutations, and RFC allele loss.


Subject(s)
Alleles , Drug Resistance, Neoplasm/genetics , Gene Silencing , Leukemia/genetics , Loss of Heterozygosity/genetics , Membrane Transport Proteins/genetics , Biological Transport/drug effects , Biological Transport/genetics , Cell Line, Tumor , Down-Regulation/drug effects , Down-Regulation/genetics , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Leukemic/genetics , Gene Silencing/drug effects , Humans , Leukemia/drug therapy , Leukemia/metabolism , Membrane Transport Proteins/metabolism , Methotrexate/pharmacology , Ornithine/analogs & derivatives , Ornithine/pharmacology , Ornithine/therapeutic use , Pterins/pharmacology , Pterins/therapeutic use , Reduced Folate Carrier Protein , Response Elements/drug effects , Response Elements/genetics , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Cancer Res ; 65(23): 10952-8, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16322243

ABSTRACT

Overexpression of the multidrug efflux transporter ABCG2 in the plasma membrane of cancer cells confers resistance to various anticancer drugs, including mitoxantrone. Here, we explored the mechanism underlying drug resistance in the MCF-7 breast cancer sublines MCF-7/MR and MCF-7/FLV1000 cells in which wild-type (R482) ABCG2 overexpression is highly confined to cell-cell attachment zones. The latter comprised the membrane of novel extracellular vesicles in which mitoxantrone was rapidly and dramatically sequestered. After 12 hours of incubation with mitoxantrone, the estimated intravesicular drug concentration was approximately 1,000-fold higher than in the culture medium. This drug compartmentalization was prevented by the specific and potent ABCG2 transport inhibitors Ko143 and fumitremorgin C, thereby resulting in restoration of drug sensitivity. Consistently, this intravesicular drug concentration was abrogated by energy deprivation and was restored upon provision of energy substrates. Fine-structure studies corroborated the presence of numerous large extracellular vesicles that were highly confined to cell-cell attachment zones between neighbor cells. Furthermore, high-resolution electron microscopy revealed that the membrane of these extracellular vesicles contained microvilli-like invaginations protruding into the intravesicular lumen. It is likely that these microvilli-like projections increase the vesicular membrane surface, thereby allowing for a more efficient ABCG2-dependent intravesicular anticancer drug concentration. Hence, these novel extracellular vesicles mediate the ABCG2-dependent extraction of intracellular drug, thereby serving as cytotoxic drug disposal chambers shared by multiple neighbor cancer cells. This constitutes a novel modality of anticancer drug resistance.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Breast Neoplasms/metabolism , Mitoxantrone/pharmacology , Neoplasm Proteins/metabolism , Transport Vesicles/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Adenosine Triphosphate/metabolism , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Mitoxantrone/pharmacokinetics
11.
Cancer Res ; 65(18): 8414-22, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16166320

ABSTRACT

ABCG2 is an ATP-binding cassette transporter that confers resistance to various chemotherapeutic agents. Recent studies have established that an Arg (wild-type) to Gly mutation at amino acid 482 in ABCG2 alters substrate specificity. Here, we explored the role of this G482 mutation in antifolate resistance using a clinically relevant 4-hour drug exposure. Stable transfectants overexpressing the mutant G482 transporter displayed 120-, 1,000-, and >6,250-fold resistance to the antifolates methotrexate, GW1843, and Tomudex, respectively, relative to parental human embryonic kidney cells. Moreover, although overexpressing equal transporter levels at the plasma membrane, G482-ABCG2 cells were 6-, 23-, and >521-fold more resistant to methotrexate, GW1843, and Tomudex, respectively, than R482-ABCG2 cells. In contrast, upon a continuous (72-hour) drug exposure, both the G482- and R482-ABCG2 cells lost almost all their antifolate resistance; this result was consistent with the inability of ABCG2 to extrude long-chain antifolate polyglutamates. Ko143, a specific and potent ABCG2 inhibitor reversed methotrexate resistance in both G482- and R482-ABCG2 cells. Consistently, whereas the pool of free methotrexate in parental human embryonic kidney cells was prominent after 4 hours of transport with 1 micromol/L [3H]methotrexate, in R482- and G482-ABCG2 cells, it was minimal. Furthermore, G482-ABCG2 cells contained marked decreases in the di- and triglutamate species of [3H]methotrexate at 4 hours of incubation with methotrexate and in the tetra- and pentaglutamates at 24 hours. These changes were not associated with any significant decrease in folylypoly-gamma-glutamate synthetase activity. These results provide the first evidence that the G482-ABCG2 mutation confers high-level resistance to various hydrophilic antifolates.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Folic Acid Antagonists/pharmacology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/biosynthesis , Cell Line , Cell Membrane/metabolism , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Folic Acid Antagonists/pharmacokinetics , Humans , Methotrexate/analogs & derivatives , Methotrexate/pharmacokinetics , Methotrexate/pharmacology , Mutation , Neoplasm Proteins/biosynthesis , Polyglutamic Acid/analogs & derivatives , Polyglutamic Acid/pharmacokinetics , Polyglutamic Acid/pharmacology , Subcellular Fractions/metabolism , Transfection , Tritium
12.
Mol Pharmacol ; 67(4): 1349-59, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15657365

ABSTRACT

The unique capability of breast cancer resistance protein (BCRP/ABCG2) to export mono-, di-, and triglutamates of folates should limit cellular proliferation under conditions of folate deprivation, particularly upon BCRP overexpression. Here, we explored the mode of adaptation of BCRP-overexpressing cells to short-term folate deprivation. MCF-7/MR cells grown in high folate medium (2.3 muM folic acid) containing mitoxantrone had 62% of their overexpressed BCRP in the plasma membrane and only 38% in the cytoplasm. In contrast, cells grown for 2 weeks in folic acid-free medium followed by an adaptation week in low folate medium (1 nM folic acid) had 86% of BCRP in the cytoplasm and only 14% in the plasma membrane. Unlike BCRP, various transmembrane proteins retained their normal plasma membrane localization in folate-deprived cells. Folate deprivation was also associated with a 3-fold decrease in BCRP and multidrug resistance protein 1 (MRP1/ABCC1) levels. Confocal microscopy with folate-deprived cells revealed that cytoplasmic BCRP colocalized with calnexin, an established endoplasmic reticulum resident. The loss of BCRP from the plasma membrane in folate-deprived cells consistently resulted in a 4.5-fold increase in [(3)H]folic acid accumulation relative to MCF-7/MR cells. Hence, cellular adaptation to shortterm folate deprivation results in a selective confinement of BCRP to the cytoplasm along with a moderate decrease in BCRP and MRP1 levels aimed at preserving the poor intracellular folate pools. These results constitute a novel mechanism of cellular adaptation to short-term folate deprivation and provide further support to the possible role of BCRP in the maintenance of cellular folate homeostasis.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Breast Neoplasms/metabolism , Cytoplasm/metabolism , Folic Acid Deficiency/metabolism , Neoplasm Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Adaptation, Physiological , Cell Line, Tumor , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Female , Folic Acid/metabolism , Glycosylation , Humans , Membrane Proteins/metabolism
13.
Arthritis Rheum ; 50(7): 2130-9, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15248210

ABSTRACT

OBJECTIVE: To investigate whether interactions of sulfasalazine (SSZ) with reduced folate carrier (RFC), the dominant cell membrane transporter for natural folates and methotrexate (MTX), may limit the efficacy of combination therapy with MTX and SSZ in patients with rheumatoid arthritis. METHODS: Human RFC-(over)expressing CEM cells of T cell origin were used to analyze the effect of SSZ on the RFC-mediated cellular uptake of radiolabeled MTX and the natural folate leucovorin. Moreover, both cells with and those without acquired resistance to SSZ were used to assess the antiproliferative effects of MTX in combination with SSZ. RESULTS: Transport kinetic analyses revealed that SSZ was a potent noncompetitive inhibitor of RFC-mediated cellular uptake of MTX and leucovorin, with mean +/- SD K(i) (50% inhibitory concentration) values of 36 +/- 6 microM and 74 +/- 7 microM, respectively. Consistent with the inhibitory interaction of SSZ with RFC, a marked loss of MTX efficacy was observed when MTX was coadministered with SSZ: up to 3.5-fold for CEM cells in the presence of 0.25 mM of SSZ, and >400-fold for SSZ-resistant cells in the presence of 2.5 mM of SSZ. Importantly, along with diminished efficacy of MTX, evidence for cellular folate depletion was obtained by the demonstration of an SSZ dose-dependent decrease in leucovorin accumulation. CONCLUSION: At clinically relevant plasma concentrations, interactions of SSZ with RFC provide a biochemical rationale for 2 important clinical observations: 1) the onset of (sub)clinical folate deficiency during SSZ treatment, and 2) the lack of additivity/synergism of the combination of SSZ and MTX when these disease-modifying antirheumatic drugs are administered simultaneously. Thus, when considering use of these drugs in combination therapies, the present results provide a rationale both for the use of folate supplementation and for spacing administration of these drugs over time.


Subject(s)
Antirheumatic Agents/pharmacology , Membrane Transport Modulators , Membrane Transport Proteins/antagonists & inhibitors , Sulfasalazine/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antirheumatic Agents/administration & dosage , Antirheumatic Agents/pharmacokinetics , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Biological Transport/drug effects , Cell Line , Dose-Response Relationship, Drug , Drug Interactions , Drug Resistance , Drug Therapy, Combination , Folic Acid/metabolism , Humans , Leucovorin/pharmacokinetics , Membrane Transport Proteins/metabolism , Methotrexate/pharmacokinetics , Methotrexate/therapeutic use , Reduced Folate Carrier Protein , Sulfasalazine/administration & dosage , Sulfasalazine/therapeutic use
14.
J Biol Chem ; 279(24): 25527-34, 2004 Jun 11.
Article in English | MEDLINE | ID: mdl-15047700

ABSTRACT

Breast cancer resistance protein (BCRP/ABCG2) is currently the only ABC transporter that exports mono- and polyglutamates of folates and methotrexate (MTX). Here we explored the relationship between cellular folate status and BCRP expression. Toward this end, MCF-7 breast cancer cells, with low BCRP and moderate multidrug resistance protein 1 (MRP1/ABCC1) levels, and their mitoxantrone (MR)-resistant MCF-7/MR subline, with BCRP overexpression and low MRP1 levels, were gradually deprived of folic acid from 2.3 microm to 3 nm resulting in the sublines MCF-7/LF and MCF-7/MR-LF. These cell lines expressed only residual BCRP mRNA and protein levels and retained a poor MRP2 (ABCC2) through MRP5 (ABCC5) expression. Furthermore, MCF-7/MR-LF cells also displayed 5-fold decreased MRP1 levels relative to MCF-7/MR cells. In contrast, BCRP overexpression was largely retained in MCF-7/MR cells grown in MR-free medium containing 2.3 microm folic acid. Loss of BCRP expression in MCF-7/LF and MCF-7/MR-LF cells resulted in the following: (a) a prominent decrease in the efflux of Hoechst 33342, a BCRP substrate; (b) an approximately 2-fold increase in MR accumulation as revealed by flow cytometry; this was accompanied by a 2.5- and approximately 84-fold increased MR sensitivity in these cell lines, respectively. Consistently, Ko143, a specific BCRP inhibitor, rendered MCF-7 and MCF-7/MR cells 2.1- and approximately 16.4-fold more sensitive to MR, respectively. Loss of BCRP expression also resulted in the following: (c) an identical MTX sensitivity in these cell lines thereby losing the approximately 28-fold MTX resistance of the MCF-7/MR cells; (d) an approximately 2-fold increase in the 4- and 24-h accumulation of [(3)H]folic acid. Furthermore, MCF-7/MR-LF cells displayed a significant increase in folylpoly-gamma-glutamate synthetase activity. Hence, consistent with the mono- and polyglutamate folate exporter function of BCRP, down-regulation of BCRP and increased folylpoly-gamma-glutamate synthetase activity appear to be crucial components of cellular adaptation to folate deficiency conditions. This is the first evidence for the possible role of BCRP in the maintenance of cellular folate homeostasis.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Breast Neoplasms/drug therapy , Folic Acid/metabolism , Neoplasm Proteins/physiology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/analysis , ATP-Binding Cassette Transporters/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Flow Cytometry , Homeostasis , Humans , Immunohistochemistry , Membrane Transport Proteins/physiology , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/physiology , Neoplasm Proteins/analysis , Neoplasm Proteins/genetics , Peptide Synthases/metabolism , Reverse Transcriptase Polymerase Chain Reaction
15.
Cancer ; 100(4): 773-82, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14770434

ABSTRACT

BACKGROUND: Although the majority of children with acute lymphoblastic leukemia (ALL) are cured with combination chemotherapy containing methotrexate (MTX), drug resistance contributes to treatment failure for a substantial fraction of patients. The primary transporter for folates and MTX is the reduced folate carrier (RFC). Impaired drug transport is a documented mechanism of MTX resistance in patients with ALL; however, to the authors' knowledge it is not known whether inactivating RFC mutations are a contributing factor. METHODS: The authors devised a genomic polymerase chain reaction-single strand conformational polymorphism assay followed by sequencing and screened the entire RFC coding region for sequence alterations in DNA from 246 leukemia specimens from patients with diverse ethnic variation, 24 at the time of recurrence and the rest at the time of diagnosis. This cohort was comprised of 203 B-precursor ALL specimens (82.5%), 32 T-lineage ALL specimens (13%), and 11 acute myeloblastic leukemia specimens (4.5%). RESULTS: Of 246 DNA samples, only 3 diagnosis B-precursor ALL specimens (1.2%) were found to harbor alterations in the RFC gene, including heterozygous single nucleotide changes resulting in D56H and D522N substitutions in the first extracellular loop and the C-terminus of this transporter, respectively. The third sample had a sequence alteration in exon 3 that could not be identified because of the lack of availability of DNA. CONCLUSIONS: Whereas inactivating RFC mutations are a frequent mechanism of MTX resistance in human leukemia cell lines and in patients with osteosarcoma, they are not common and do not appear to play any significant role in intrinsic or acquired resistance to MTX in childhood leukemia. This is the first study of RFC mutations in multiple pediatric leukemia specimens.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Carrier Proteins/genetics , Carrier Proteins/pharmacology , Membrane Transport Proteins , Methotrexate/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Blotting, Northern , Child , DNA Mutational Analysis , Drug Resistance, Neoplasm , Humans , Osteosarcoma/drug therapy , Osteosarcoma/genetics , Polymerase Chain Reaction , Polymorphism, Genetic , Polymorphism, Single-Stranded Conformational , Reduced Folate Carrier Protein , Tumor Cells, Cultured
16.
Cancer ; 98(9): 1958-66, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14584080

ABSTRACT

BACKGROUND: High-dose methotrexate (MTX) is an important component of current protocols for the treatment of osteosarcoma. Although MTX uptake proceeds primarily through the reduced folate carrier (RFC) protein and efflux occurs via multidrug resistance protein 1 (MRP1), RFC protein expression in osteosarcoma remains unexamined. METHODS: RFC and MRP1 expression (normalized to beta-actin expression) was examined with Western blot analysis in 11 osteosarcoma specimens obtained at diagnosis and 9 osteosarcoma specimens obtained on recurrence. RESULTS: The average RFC level in specimens obtained on recurrence was significantly higher than the level in specimens obtained at diagnosis (P = 0.0005). Furthermore, in all three matched pairs of diagnosis and recurrence specimens, RFC levels were higher in recurrence specimens than in the corresponding diagnosis specimens. Potential correlations between RFC and MRP1 expression and histologic response to preoperative chemotherapy were examined. Tumors with poor histologic responses (i.e.,

Subject(s)
Bone Neoplasms/metabolism , Carrier Proteins/metabolism , Membrane Transport Proteins , Osteosarcoma/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , Actins/analysis , Adolescent , Adult , Bone Neoplasms/drug therapy , Child , Drug Resistance, Neoplasm , Humans , Male , Membrane Proteins/analysis , Methotrexate/therapeutic use , Middle Aged , Neoplasm Recurrence, Local , Osteosarcoma/drug therapy , Reduced Folate Carrier Protein , Tumor Cells, Cultured
17.
J Biol Chem ; 278(9): 6680-6, 2003 Feb 28.
Article in English | MEDLINE | ID: mdl-12486126

ABSTRACT

We studied the molecular basis of the up to 46-fold increased accumulation of folates and methotrexate (MTX) in human leukemia CEM-7A cells established by gradual deprivation of leucovorin (LCV). CEM-7A cells consequently exhibited 10- and 68-fold decreased LCV and folic acid growth requirements and 23-25-fold hypersensitivity to MTX and edatrexate. Although CEM-7A cells displayed a 74-86-fold increase in the reduced folate carrier (RFC)-mediated influx of LCV and MTX, RFC overexpression per se cannot induce a prominently increased folate/MTX accumulation because RFC functions as a nonconcentrative anion exchanger. We therefore explored the possibility that folate efflux activity mediated by members of the multidrug resistance protein (MRP) family was impaired in CEM-7A cells. Parental CEM cells expressed substantial levels of MRP1, MRP4, poor MRP5 levels, whereas MRP2, MRP3 and breast cancer resistance protein were undetectable. In contrast, CEM-7A cells lost 95% of MRP1 levels while retaining parental expression of MRP4 and MRP5. Consequently, CEM-7A cells displayed a 5-fold decrease in the [(3)H]folic acid efflux rate constant, which was identical to that obtained with parental CEM cells, when their folic acid efflux was blocked (78%) with probenecid. Furthermore, when compared with parental CEM, CEM-7A cells accumulated 2-fold more calcein fluorescence. Treatment of parental cells with the MRP1 efflux inhibitors MK571 and probenecid resulted in a 60-100% increase in calcein fluorescence. In contrast, these inhibitors failed to alter the calcein fluorescence in CEM-7A cells, which markedly lost MRP1 expression. Replenishment of LCV in the growth medium of CEM-7A cells resulted in resumption of normal MRP1 expression. These results establish for the first time that MRP1 is the primary folate efflux route in CEM leukemia cells and that the loss of folate efflux activity is an efficient means of markedly augmenting cellular folate pools. These findings suggest a functional role for MRP1 in the maintenance of cellular folate homeostasis.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Folic Acid/metabolism , Biological Transport , Blotting, Western , Cell Division , Cell Line , Cell Membrane/metabolism , Flow Cytometry , Fluoresceins/pharmacology , Folic Acid/pharmacology , Humans , Leucovorin/pharmacology , Methotrexate/pharmacology , Microscopy, Fluorescence , Mutation , Time Factors , Tumor Cells, Cultured
18.
Biochem J ; 367(Pt 3): 741-50, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12139489

ABSTRACT

We have studied the molecular basis of resistance of multiple human leukaemia CCRF-CEM sublines to the novel antifolates ZD9331, GW1843, AG2034, PT523 and edatrexate, which use the reduced folate carrier (RFC) as their main cellular uptake route and that target different folate-dependent enzymes. Antifolate-resistant sublines established by stepwise and single-step selections displayed up to 2135-fold resistance to the selection drug, and up to 2323-fold cross-resistance to various hydrophilic antifolates. In contrast, these sublines were up to 17- and 20-fold hypersensitive to the lipophilic antifolates AG377 and trimetrexate, respectively. The total reduced folate pool of these antifolate-resistant sublines shrunk by 87-96%, resulting in up to 42-fold increased folic acid growth requirement. These sublines lost 92-97% of parental [(3)H]methotrexate influx rates. Genomic PCR single-strand conformational polymorphism analysis and sequencing revealed that most of these drug-resistant sublines harboured RFC mutations that surprisingly clustered in two confined regions in exons 2 and 3. The majority of these mutations resulted in frame-shift and/or premature translation termination and lack of RFC protein expression. The remaining mutations involved single amino acid substitutions predominantly residing in the first transmembrane domain (TMD1). Some RFC-inactivating mutations emerged during the early stages of antifolate selection and were stably retained during further drug selection. Furthermore, some sublines displayed a markedly decreased or abolished RFC mRNA and/or protein expression. This constitutes the first demonstration of clustering of multiple human RFC mutations in TMD1, thereby suggesting that it plays a functional role in folate/antifolate binding and/or translocation. This is the first molecular characterization of human RFC-associated modalities of resistance to various novel antifolates in multiple leukaemia sublines.


Subject(s)
Carrier Proteins/genetics , Drug Resistance, Neoplasm/genetics , Folic Acid Antagonists/pharmacology , Leukemia/metabolism , Membrane Transport Proteins , Mutation , Base Sequence , Blotting, Western , DNA Primers , Folic Acid Antagonists/pharmacokinetics , Humans , Leukemia/pathology , Methotrexate/pharmacokinetics , Methotrexate/pharmacology , Polymorphism, Single-Stranded Conformational , Reduced Folate Carrier Protein , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...