Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Cancer Res ; 61(19): 7225-32, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11585759

ABSTRACT

We have recently determined that human multidrug resistance protein (MRP) 3, which confers resistance to certain natural product agents and methotrexate (MTX), is competent in the MgATP-energized transport of MTX and the monoanionic bile constituent glycocholate as well as several glutathione and glucuronate conjugates. Of these capabilities, the facility of MRP3 in conferring resistance to and mediating the transport of MTX is of particular interest because it raises the possibility that this pump is a component of the previously described cellular efflux system for this antimetabolite. However, if this is to be the case, a critical property of cellular MTX efflux that must be addressed is its ability to mediate the export of MTX but not that of its intracellular polyglutamylated derivatives. Here we examine the role of MRP3 in these and related processes by determining the selectivity of this transporter for MTX, MTX polyglutamates, and physiological folates. In so doing, we show that MRP3 is not only active in the transport of MTX but is also active in the transport the physiological folates folic acid (FA) and N(5)-formyltetrahydrofolic acid (leucovorin) and that polyglutamylation of MTX abolishes transport. Both FA and leucovorin are subject to high-capacity (V(max(FA)), 1.71 +/- 0.05 nmol/mg/min; V(max(leucovorin)), 3.63 +/- 1.20 nmol/mg/min), low-affinity (K(m(FA)), 1.96 +/- 0.13 mM; K(m(leucovorin)), 1.74 +/- 0.65 mM) transport by MRP3. Addition of a single glutamyl residue to MTX is sufficient to diminish transport by >95%. We also show that polyglutamylation similarly affects the capacity of MRP1 to transport MTX and that physiological folates are also subject to MgATP-stimulated transport by MRP1. On the basis of the capacity to transport MTX but not MTX-Glu(2), it is concluded that MRP3 and MRP1 represent components of the previously described cellular efflux system for MTX. The capacity of MRP3 to transport folates indicates that it may reduce intracellular levels of these compounds and thereby indirectly influence antifolate cytotoxicity, and it also implies that this pump may play a role in the response to chemotherapeutic regimens in which leucovorin is a component.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Drug Resistance, Multiple , Folic Acid/pharmacokinetics , Leucovorin/pharmacokinetics , Methotrexate/pharmacokinetics , Polyglutamic Acid/pharmacokinetics , 3T3 Cells , ATP-Binding Cassette Transporters/antagonists & inhibitors , Animals , Biological Transport/drug effects , Humans , Methotrexate/analogs & derivatives , Methotrexate/metabolism , Mice , Multidrug Resistance-Associated Proteins , Osmolar Concentration , Polyglutamic Acid/analogs & derivatives , Polyglutamic Acid/metabolism , Tritium
2.
J Biol Chem ; 276(36): 33747-54, 2001 Sep 07.
Article in English | MEDLINE | ID: mdl-11447229

ABSTRACT

Human multidrug resistance protein 4 (MRP4) has recently been determined to confer resistance to the antiviral purine analog 9-(2-phosphonylmethoxyethyl)adenine and methotrexate. However, neither its substrate selectivity nor physiological functions have been determined. Here we report the results of investigations of the in vitro transport properties of MRP4 using membrane vesicles prepared from insect cells infected with MRP4 baculovirus. It is shown that expression of MRP4 is specifically associated with the MgATP-dependent transport of cGMP, cAMP, and estradiol 17-beta-D-glucuronide (E(2)17 beta G). cGMP, cAMP, and E(2)17 beta G are transported with K(m) and V(max) values of 9.7 +/- 2.3 microm and 2.0 +/- 0.3 pmol/mg/min, 44.5 +/- 5.8 microm and 4.1 +/- 0.4 pmol/mg/min, and 30.3 +/- 6.2 microm and 102 +/- 16 pmol/mg/min, respectively. Consistent with its ability to transport cyclic nucleotides, it is demonstrated that the MRP4 drug resistance profile extends to 6-mercaptopurine and 6-thioguanine, two anticancer purine analogs that are converted in the cell to nucleotide analogs. On the basis of its capacity to transport cyclic nucleotides and E(2)17 beta G, it is concluded that MRP4 may influence diverse cellular processes regulated by cAMP and cGMP and that its substrate range is distinct from that of any other characterized MRP family member.


Subject(s)
Carrier Proteins/chemistry , Drug Resistance , Estradiol/chemistry , Mercaptopurine/pharmacology , Thioguanine/pharmacology , 3T3 Cells , Animals , Anion Transport Proteins , Antimetabolites, Antineoplastic/pharmacology , Baculoviridae/metabolism , Biological Transport , Cell Membrane/metabolism , Cells, Cultured , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Dose-Response Relationship, Drug , Estradiol/analogs & derivatives , Immunoblotting , Insecta , Kinetics , Mice , Models, Biological , Osmosis , Time Factors , Transfection
3.
Int J Cancer ; 93(1): 107-13, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11391629

ABSTRACT

We previously isolated agosterol A (AG-A) from a marine Spongia sp. and found that it completely reversed colchicine resistance in P-glycoprotein (Pgp)-over-expressing KB-C2 cells and vincristine resistance in multidrug-resistance protein (MRP)1-over-expressing CV60 cells. However, a tri-deacetylated derivative of AG-A (IAG-A) showed almost no activity in reversing Pgp- or MRP1-mediated drug resistance. In this study, we examined the mechanisms by which AG-A reverses MRP1-mediated drug resistance by investigating the interaction between agosterols and MRP1 in MRP1-over-expressing human KB carcinoma (KB/MRP) cells. [3H]-Leukotriene C4 (LTC4), [3H]-2,4-dinitrophenyl-S-glutathione uptake into membrane vesicles prepared from KB/MRP cells and intracellular [3H]-vincristine accumulation and efflux in KB/MRP cells were measured with or without AG-A and/or inactive IAG-A. AG-A reduced MRP1-mediated [3H]-LTC4 transport in a dose-dependent manner, but IAG-A did not. Inhibition by AG-A was competitive, with a K(i) value of 31 microM. AG-A at 10 microM enhanced the accumulation of [3H]-vincristine in KB/MRP cells to the level of that in control cells in the absence of the agent. Likewise, ATP-dependent efflux of [3H]-vincristine from KB/MRP cells was enhanced compared with KB-3-1 cells and inhibited by AG-A. In addition, AG-A reduced intracellular levels of glutathione, a compound required for MRP1-mediated transport of some anti-cancer drugs. These findings suggest that AG-A reverses MRP1-mediated drug resistance by directly inhibiting the capacity of MRP1 to transport drugs. In addition, the capacity of AG-A to reduce cellular glutathione levels may contribute to the modulating activity of MRP1.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , Antineoplastic Agents/toxicity , Colchicine/pharmacology , Drug Resistance, Multiple , Marine Toxins/toxicity , Sterols/toxicity , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adenosine Triphosphate/metabolism , Biological Transport/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Survival/drug effects , Doxorubicin/toxicity , Etoposide/toxicity , Gene Expression Regulation/drug effects , Humans , KB Cells , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Vincristine/pharmacokinetics , Vincristine/toxicity
4.
Cancer Lett ; 162(2): 181-91, 2001 Jan 26.
Article in English | MEDLINE | ID: mdl-11146224

ABSTRACT

The MRP subfamily of ABC transporters currently consists of at least six members, several of which have been demonstrated to transport amphipathic anions and to confer in vitro resistance to chemotherapeutic agents. In searching the data bases we identified the product of a cDNA sequencing project that bears significant similarity to MRP subfamily transporters. In this report the predicted coding sequence, protein product and expression pattern of this cDNA, termed MRP7, are analyzed. The MRP7 cDNA sequence encodes a 1492 amino acid ABC transporter whose structural architecture resembles that of MRP1, MRP2, MRP3, and MRP6, in that its transmembrane helices are arranged in three membrane spanning domains. However, in contrast to the latter transporters, a conserved N-linked glycosylation site is not found at the N-terminus of MRP7. Comparisons of the MRP7 amino acid sequence indicated that while it is most closely related to other MRP subfamily members, its degree of relatedness is the lowest of any of the known MRP-related transporters. The integrity of the predicted MRP7 coding sequence was confirmed by the synthesis of an approximately 158 kDa protein in reticulocyte lysates programmed with the MRP7 cDNA. While MRP7 transcript was detected in a variety of tissues by RT/PCR, it was not readily detectable by RNA blot analysis, suggesting that it is expressed at low levels in these tissues. Fluorescence in situ hybridization indicated that MRP7 maps to chromosome 6p12-21, in proximity to several genes associated with glutathione conjugation and synthesis. On the basis of these findings and evolutionary cluster analysis, we conclude that MRP7 is a member of the MRP subfamily of amphipathic anion transporters.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/chemistry , Amino Acid Sequence , Cluster Analysis , DNA, Complementary/genetics , DNA, Complementary/metabolism , Gene Expression Profiling , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Multidrug Resistance-Associated Proteins , Protein Structure, Tertiary , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
5.
J Bioenerg Biomembr ; 33(6): 493-501, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11804191

ABSTRACT

The MRP subfamily of ABC transporters from mammals consists of at least seven members, six of which have been implicated in the transport of amphipathic anions. MRP1, MRP2, and MRP3 bear a close structural resemblance, confer resistance to a variety of natural products as well as methotrexate, and have the facility for transporting glutathione and glucuronate conjugates. MRP1 is a ubiquitously expressed efflux pump for the products of phase II of xenobiotic detoxification, while MRP2, whose hereditary deficiency results in Dubin-Johnson syndrome, functions to extrude organic anions into the bile. MRP3 is distinguished by its capacity to transport the monoanionic bile constituent glycocholate, and may function as a basolateral back-up system for the detoxification of hepatocytes when the usual canalicular route is impaired by cholestatic conditions. MRP4 and MRP5 resemble each other more closely than they resemble MRPs 1-3 and confer resistance to purine and nucleotide analogs which are either inherently anionic, as in the case of the anti-AIDS drug PMEA, or are phosphorylated and converted to anionic amphiphiles in the cell, as in the case of 6-MP. Given their capacity for transporting cyclic nucleotides, MRP4 and MRP5 have also been implicated in a broad range of cellular signaling processes. The drug resistance activity and physiological substrates of MRP6 are unknown. However, its hereditary deficiency results in pseudoxanthoma elasticum, a multisystem disorder affecting skin, eyes, and blood vessels. It is hoped that elucidation of the resistance profiles and physiological functions of the different members of the MRP subfamily will provide new insights into the molecular basis of clinical drug resistance and spawn new strategies for combating this phenomenon.


Subject(s)
Drug Resistance, Neoplasm/physiology , Multidrug Resistance-Associated Proteins/physiology , Amino Acids/chemistry , Animals , Humans , Phenotype
6.
J Natl Cancer Inst ; 92(23): 1934-40, 2000 Dec 06.
Article in English | MEDLINE | ID: mdl-11106685

ABSTRACT

BACKGROUND: Multidrug resistance-associated protein (MRP) 1 and canalicular multispecific organic anion transporter (cMOAT or MRP2) are adenosine triphosphate-binding cassette transporters that confer resistance to anticancer agents. In addition to these two transporters, there are at least four other human MRP subfamily members (MRP3 through MRP6). We and others reported previously that MRP3 is capable of conferring resistance to certain anticancer agents. In this study, we investigated whether MRP4 (MOAT-B), whose transcript accumulates to the highest levels in prostate tissue, has the capacity to confer drug resistance. METHODS: MRP4-transfected NIH3T3 cells were generated, and their drug sensitivity was analyzed. The subcellular localization of MRP4 was assessed by immunohistochemical analysis in transfected cells and in prostate tissue. Statistical tests were two-sided. RESULTS: MRP4 was detected as a 170-kd protein that was localized in the plasma membrane and cytoplasm of transfected cells. The MRP4 transfectants displayed 5.5-fold increased resistance to methotrexate in short-term drug-exposure assays (P=.022) and exhibited decreased cellular accumulation of this agent at 4 hours (P=.006) and 24 hours (P<.001). In continuous-exposure assays, however, the MRP4 transfectants did not display increased resistance for either methotrexate or natural product cytotoxic agents (anthracyclines, etoposide, vinca alkaloids, and paclitaxel [Taxol]). However, the transfectants did show increased resistance (2.3-fold) for the anti-acquired immunodeficiency syndrome nucleoside analogue 9-(2-phosphonylmethoxyethyl)adenine (PMEA) (P=.022) in continuous-exposure assays. Consistent with MRP4's plasma membrane localization in transfected cells, analysis of prostate tissue showed that MRP4 protein was localized primarily in the basolateral plasma membranes of tubuloacinar cells. CONCLUSIONS: These results indicate that MRP4 confers resistance to short-term methotrexate and continuous PMEA treatment. Given its structure, drug resistance profile and subcellular localization, MRP4 probably functions as an amphipathic anion efflux pump whose substrate range includes glutamate and phosphate conjugates.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/genetics , Antineoplastic Agents/pharmacology , Carrier Proteins/genetics , Drug Resistance, Multiple , Drug Resistance, Neoplasm , 3T3 Cells , ATP Binding Cassette Transporter, Subfamily B/physiology , Acrylates/pharmacology , Animals , Anion Transport Proteins , Antibiotics, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Carrier Proteins/physiology , Cloning, Molecular , DNA, Complementary/genetics , Drug Screening Assays, Antitumor , Humans , Immunoblotting , Immunohistochemistry , Male , Methotrexate/pharmacology , Mice , Polymers/pharmacology , Prostate/metabolism , Transfection
7.
Cancer Res ; 60(17): 4779-84, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10987286

ABSTRACT

The multidrug resistance-associated protein 1 (MRP1) and the canalicular multispecific organic anion transporter (cMOAT or MRP2) are ATP-binding cassette transporters that confer resistance to some anticancer drugs and efflux glutathione and glucuronate conjugates from the cell. The MRP subfamily of ABC transporters, however, contains at least four other members of which MRP3 (MOAT-D) bears the closest structural resemblance to MRP1. Although transfection studies have established that human MRP3 confers increased resistance to several anticancer agents, neither the substrate selectivity nor physiological functions of this transporter have been determined. Here we report the results of investigations of the in vitro transport properties of cloned human MRP3 using membrane vesicles prepared from MRP3-transfected HEK293 cells. It is shown that the expression of MRP3 is specifically associated with enhancement of the MgATP-dependent transport into membrane vesicles of the glucuronide estradiol 17-beta-D-glucuronide (E(2)17betaG), the glutathione conjugates 2,4-dinitrophenyl S-glutathione (DNP-SG) and leukotriene C4 (LTC4), the antimetabolite methotrexate, and the bile acid glycocholate. DNP-SG, LTC4, and E(2)17betaG are transported at moderate affinity and low capacity with Km and Vmax values of 5.7 +/- 1.7 microM and 3.8 +/- 0.1 pmol/mg/min, 5.3 +/- 2.6 microM and 20.2 +/- 5.9 pmol/mg/min, and 25.6 +/- 5.4 microM and 75.6 +/- 5.9 pmol/mg/min, respectively. Methotrexate and glycocholate are transported at low affinity and high capacity with Km and Vmax values of 776 +/- 319 microM and 288 +/- 54 pmol/mg/min and 248 +/- 113 microM and 183 +/- 34 pmol/mg/min, respectively. On the basis of these findings, the osmotic dependence of the transport measured and its inability to transport taurocholate, MRP3, like MRP1 and cMOAT, is concluded to be competent in the transport of glutathione S-conjugates, glucuronides, and methotrexate, albeit at low to moderate affinity. In contrast to MRP1, cMOAT, and all other characterized mammalian ABC transporters, however, MRP3 is active in the transport of the monoanionic human bile constituent glycocholate.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Carrier Proteins/metabolism , Estradiol/analogs & derivatives , Glutathione/analogs & derivatives , Multidrug Resistance-Associated Proteins , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adenosine Triphosphate/metabolism , Anion Transport Proteins , Biological Transport, Active , Carbon Radioisotopes , Cell Line , Cell Membrane/metabolism , Estradiol/pharmacokinetics , Glutathione/pharmacokinetics , Glycocholic Acid/pharmacokinetics , Humans , Kidney/metabolism , Kinetics , Leukotriene C4/pharmacokinetics , Methotrexate/pharmacokinetics , Osmosis , Substrate Specificity , Tritium
8.
J Pharmacol Exp Ther ; 294(2): 480-7, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10900222

ABSTRACT

Glutathione (GSH), glutathione S-transferases (GSTs), and the multidrug resistance-associated protein 1 (MRP1) have been independently studied for their contributions to drug resistance. Single cDNA transfection experiments have provided inconsistent and disparate conclusions with respect to the importance of GSH and GST in conferring a resistant phenotype. Because these three proteins can act as a concerted coordinated pathway, we reasoned that equivalent increases may be required for enhanced resistance to be expressed. We have assembled these proteins together, or in various combinations, to determine whether they show cooperativity in determining drug response. Increased expression through single cDNA transfection of GSTpi, gamma-glutamylcysteine synthetase (gamma-GCS) (regulatory plus catalytic subunits), or MRP1 enhanced resistance to a number of anticancer drugs. Cotransfection of GSTpi and GCS, gave higher resistance to doxorubicin, etoposide, and vincristine than with either alone. Resistance toward chlorambucil and ethacrynic acid was similar in cells overexpressing either component or overexpressing GST alone. Coexpression of GSTpi with MRP1 conferred significant resistance above that seen for MRP1 alone to chlorambucil, etoposide, ethacrynic acid, and vincristine. The combination of GCS and MRP1 did not afford additional resistance above MRP1 alone. When all three were transfected, significantly higher levels of resistance were found for doxorubicin and etoposide. These results support the concept that coordinate enhancement of focal thiol elements of detoxification pathways provides a more efficient protective phenotype than do single components alone.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Drug Resistance, Neoplasm/physiology , Glutamate-Cysteine Ligase/biosynthesis , Glutathione Transferase/biosynthesis , Glutathione/metabolism , Isoenzymes/biosynthesis , 3T3 Cells , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Amino Acid Sequence , Animals , Cell Survival/drug effects , Cell Survival/physiology , DNA, Complementary/genetics , DNA, Complementary/metabolism , Drug Screening Assays, Antitumor , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/metabolism , Glutathione S-Transferase pi , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , HT29 Cells , Humans , Inactivation, Metabolic , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Molecular Sequence Data , Multidrug Resistance-Associated Proteins , Transfection
9.
Cancer Res ; 59(23): 5964-7, 1999 Dec 01.
Article in English | MEDLINE | ID: mdl-10606242

ABSTRACT

Multidrug resistance-associated protein (MRP)1 and canalicular multispecific organic anion transporter (cMOAT)/MRP2 are ATP-binding cassette (ABC) transporters that confer resistance to natural product cytotoxic drugs. We recently described the complete coding sequences of four human MRP/cMOAT subfamily members and found that, among these proteins, MRP3/MOAT-D is most closely related to MRP1 (58% identity; M. G. Belinsky and G. D. Kruh, Br. J. Cancer, 80: 1342-1349, 1999). In the present study, we sought to determine whether MRP3 is capable of conferring resistance to cytotoxic drugs. To address this question, human embryonic kidney 293 cells were transfected with an MRP3 expression vector, and the drug resistance phenotype of the transfected cells was analyzed. The MRP3-transfected cells displayed approximately 4-fold resistance to etoposide and approximately 2-fold resistance to vincristine, compared with control transfected cells. In addition, approximately 1.7-fold resistance was observed for the antimetabolite methotrexate. Increased resistance was not observed for several other natural product agents, including anthracyclines and Taxol. The MRP-transfected cells exhibited reduced accumulation of radiolabeled etoposide, consistent with the operation of a plasma membrane efflux pump. These results indicate that MRP3 confers resistance to some anticancer agents but that its resistance pattern is distinct from the resistance patterns of other ABC transporters involved in resistance to natural product chemotherapeutic agents.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Antineoplastic Agents/toxicity , Drug Resistance, Multiple , Etoposide/pharmacokinetics , Etoposide/toxicity , Multidrug Resistance-Associated Proteins , Vincristine/toxicity , ATP-Binding Cassette Transporters/genetics , Cell Line , Cell Survival/drug effects , Embryo, Mammalian , Humans , Kidney , Recombinant Proteins/metabolism , Transfection
10.
Br J Cancer ; 80(9): 1342-9, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10424734

ABSTRACT

Multidrug resistance-associated protein (MRP) and the canalicular multispecific organic anion transporter (cMOAT) are organic anion pumps that have been linked to cytotoxic drug resistance. We previously reported the isolation of three human MRP/cMOAT-related transporters, MOAT-B (MRP4), MOAT-C (MRP5) and MOAT-D (MRP3). In the present study we describe the fourth MRP/cMOAT-related transporter. We analysed ARA, a human cDNA reported to encode a 453 residue MRP-related transporter, and found that it represents a fused transcript composed of MRP sequences and partial sequences of a novel transporter. The complete coding sequence of this novel transporter, which we designated MOAT-E, was isolated. MOAT-E encodes a 1503 residue transporter that is most closely related to MRP (45%), MOAT-D (44%) and cMOAT (39%), both in terms of amino acid identity and sharing a common topology in which approximately 17 transmembrane spanning helices are distributed within three membrane spanning domains. RNA blot analysis indicated that MOAT-E expression is restricted to kidney and liver. These observations suggest that MOAT-E may function as an organic anion transporter involved in cellular detoxification and possibly in the hepatobiliary and renal excretion of xenobiotics and/or endogenous metabolites. Isolation of MOAT-E helps to define the MRP/cMOAT subfamily of transporters.


Subject(s)
ATP-Binding Cassette Transporters/analysis , Carrier Proteins/analysis , Kidney/chemistry , Liver/chemistry , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , Amino Acid Sequence , Anion Transport Proteins , Carrier Proteins/chemistry , Carrier Proteins/genetics , DNA, Complementary/isolation & purification , Humans , Molecular Sequence Data , Multidrug Resistance-Associated Proteins
11.
J Natl Cancer Inst ; 90(22): 1735-41, 1998 Nov 18.
Article in English | MEDLINE | ID: mdl-9827529

ABSTRACT

BACKGROUND: Multidrug resistance-associated protein (MRP) and canalicular multispecific organic anion transporter (cMOAT) are transporter proteins that pump organic anions across cellular membranes and have been linked to resistance to cytotoxic drugs. We previously identified MOAT-B, an MRP/cMOAT-related transporter, by use of a polymerase chain reaction approach. However, analysis of expressed sequence tag (EST) databases indicated that there might be additional MRP/cMOAT-related transporters. To further define the MRP/cMOAT subfamily of transporters, we used EST probes to isolate complementary DNAs for two related transporter proteins, MOAT-C and MOAT-D. METHODS: MOAT-C and MOAT-D expression patterns in human tissues were determined by RNA blot analysis, and chromosomal localization of the genes was determined by fluorescence in situ hybridization. RESULTS: MOAT-C is predicted to encode a 1437-amino-acid protein that, among eukaryotic transporters, is most closely related to MRP, cMOAT, and MOAT-B (about 36% identity). However, MOAT-C is less related to MRP and cMOAT than MRP and cMOAT are to each other (about 48% identity). Like MOAT-B, MOAT-C lacks an N-terminal membrane-spanning domain, indicating that the topology of this protein is similarly distinct from that of MRP and cMOAT. MOAT-D is predicted to encode a 1527-amino-acid protein that is the closest known relative of MRP (about 58% identity). MOAT-D is also highly related to cMOAT (about 47% identity). The presence of an N-terminal membrane-spanning domain indicates that the topology of MOAT-D is quite similar to that of MRP and cMOAT. MOAT-C transcripts are widely expressed in human tissues; however, MOAT-D transcript expression is more restricted. The MOAT-C and MOAT-D genes are located at chromosomes 3q27 and 17q21.3, respectively. CONCLUSIONS: On the basis of amino acid identity and protein topology, the MRP/cMOAT transporter subfamily falls into two groups; the first group consists of MRP, cMOAT, and MOAT-D, and the second group consists of MOAT-B and MOAT-C.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , Carrier Proteins/chemistry , DNA, Neoplasm/analysis , Leukemia/genetics , Neoplasm Proteins/chemistry , Ovarian Neoplasms/genetics , ATP-Binding Cassette Transporters/genetics , Amino Acid Sequence , Anion Transport Proteins , Carrier Proteins/genetics , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 3/genetics , DNA, Neoplasm/isolation & purification , Female , Gene Expression Regulation, Neoplastic , Humans , Molecular Sequence Data , Multidrug Resistance-Associated Proteins , Neoplasm Proteins/genetics , Sequence Analysis, DNA
12.
Biochim Biophys Acta ; 1381(2): 131-8, 1998 Jul 23.
Article in English | MEDLINE | ID: mdl-9685611

ABSTRACT

Acquired resistance to adriamycin (ADR) in an HL60 cell line is shown to be accompanied by an increase in DNA-dependent protein kinase catalytic subunit (DNA-PKcs) at both the protein and mRNA levels (15-20-fold) and an overall 3-fold increase in DNA-PK enzyme activity. The other components of the DNA-PK Ku autoantigen complex, Ku70 and Ku80, were 3-fold increased and unchanged, respectively. Time dependent repair of ADR-induced DNA damage was measured by the neutral comet assay and found to be more efficient in the drug resistant cell line (HL60/ADR). Antisense RNA transfection reduced the protein expression of DNA-PKcs to 50% in HL60/ADR and partially reversed drug resistance. A fibroblast cell line from a severe combined immunodeficient (SCID) mouse was deficient in functional DNA-PKcs and showed increased sensitivity to ADR and other DNA damaging agents compared to wild type. These studies demonstrate that alteration in DNA-PK can contribute to chronic stress response leading to acquired drug resistance. The overexpression of DNA-PK is thus shown to be a novel cellular adaptation mechanistically contributing to the resistance of cancer cells to the anthracycline drug adriamycin, and as such, may have implications for its therapeutic use.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antigens, Nuclear , DNA Helicases , Doxorubicin/pharmacology , Protein Serine-Threonine Kinases/metabolism , Adaptation, Physiological , Animals , Base Sequence , Cell Line , DNA Damage , DNA Primers/genetics , DNA Repair , DNA-Activated Protein Kinase , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drug Resistance/genetics , Drug Resistance/physiology , Gene Expression , HL-60 Cells , Humans , Ku Autoantigen , Mice , Mice, SCID , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Polymerase Chain Reaction , Protein Conformation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , RNA, Antisense/genetics , Transfection
13.
Cancer Res ; 58(13): 2741-7, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9661885

ABSTRACT

Multidrug resistance-associated protein (MRP) and canalicular multispecific organic anion transporter (cMOAT) are closely related mammalian ATP-binding cassette transporters that export organic anions from cells. Transfection studies have established that MRP confers resistance to natural product cytotoxic agents, and recent evidence suggests the possibility that cMOAT may contribute to cytotoxic drug resistance as well. Based upon the potential importance of these transporters in clinical drug resistance and their important physiological roles in the export of the amphiphilic products of phase I and phase II metabolism, we sought to identify other MRP-related transporters. Using a degenerate PCR approach, we isolated a cDNA that encodes a novel ATP-binding cassette transporter, which we designated MOAT-B. The MOAT-B gene was mapped using fluorescence in situ hybridization to chromosome band 13q32. Comparison of the MOAT-B predicted protein with other transporters revealed that it is most closely related to MRP, cMOAT, and the yeast organic anion transporter YCF1. Although MOAT-B is closely related to these transporters, it is distinguished by the absence of a approximately 200 amino acid NH2-terminal hydrophobic extension that is present in MRP and cMOAT and which is predicted to encode several transmembrane spanning segments. In addition, the MOAT-B tissue distribution is distinct from MRP and cMOAT. In contrast to MRP, which is widely expressed in tissues, including liver, and cMOAT, the expression of which is largely restricted to liver, the MOAT-B transcript is widely expressed, with particularly high levels in prostate, but is barely detectable in liver. These data indicate that MOAT-B is a ubiquitously expressed transporter that is closely related to MRP and cMOAT and raise the possibility that it may be an organic anion pump relevant to cellular detoxification.


Subject(s)
Carrier Proteins/chemistry , Chromosomes, Human, Pair 13 , Drug Resistance, Multiple , ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , Amino Acid Sequence , Anion Transport Proteins , Base Sequence , Carrier Proteins/genetics , Chromosome Mapping , Chromosomes, Human, Pair 13/genetics , Drug Resistance, Multiple/genetics , Humans , Molecular Sequence Data
14.
Oncogene ; 16(15): 1903-12, 1998 Apr 16.
Article in English | MEDLINE | ID: mdl-9591773

ABSTRACT

Many Src Homology 3 (SH3) domains function as molecular adhesives in intracellular signal transduction. Based on previous ultrastructural studies, short motifs which bind to the first SH3 domains of the adapters Crk and CRKL were selectively mutagenised to generate Crk/CRKL SH3-binding peptides of very high affinity and selectivity. Affinities were increased up to 20-fold compared to the best wildtype sequences, while the selectivity against a similar SH3 domain [Grb2SH3(N)] was not only retained, but sometimes increased. Blot techniques with GST-fusion peptides and in solution precipitation assays with biotinylated high affinity Crk binding peptides (HACBPs) were subsequently used to analyse the binding of these sequences to a large panel of SH3 domain-containing fusion proteins. Only those proteins which contained the CrkSH3(1) or CRKLSH3(1) domains bound efficiently to the HACBPs. A GST-HACBP fusion protein precipitated Crk and CRKL proteins out of 35S-labelled and unlabelled cell lysates. Very little binding of other cellular proteins to HACBP was detectable, indicative of a great preference for Crk and CRKL when compared to the wide variety of other endogenous cellular proteins. Moreover, HACBP disrupted in vitro preexisting Crk-complexes with DOCK180 and the exchange factors SoS and C3G, which are known targets of Crk adapters, in a concentration dependent manner. HACBP-based molecules should therefore be useful as highly selective inhibitors of intracellular signalling processes involving Crk and CRKL.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , rac GTP-Binding Proteins , src Homology Domains , Animals , Blotting, Western , Guanine Nucleotide Exchange Factors , HeLa Cells , Humans , Mice , Proto-Oncogene Proteins c-crk , Son of Sevenless Proteins
15.
Cancer Res ; 58(7): 1332-7, 1998 Apr 01.
Article in English | MEDLINE | ID: mdl-9537224

ABSTRACT

An estramustine-resistant human ovarian carcinoma cell line, SKEM, was generated to explore resistance mechanisms associated with this agent. Cytogenetic analysis revealed that SKEM cells have a homogeneously staining region (hsr) at chromosome 9q34. Microdissection of the hsr, followed by fluorescence in situ hybridization to SKEM and normal metaphase spreads, confirmed that the amplified region was derived from sequences from 9q34. In situ hybridization with a probe specific for ABC2, a gene located at 9q34 that encodes an ATP-binding cassette 2 (ABC2) transporter, indicated that this gene is amplified approximately 6-fold in the estramustine-resistant cells. Southern analysis confirmed that ABC2 was amplified in SKEM, and Northern analysis indicated that the ABC2 transcript was overexpressed approximately 5-fold. The ABC1 gene located at 9q22-31 was not amplified in the resistant cells, and mRNA levels of several other ABC transporter genes were unaltered. Consistent with the concept that increased ABC2 expression contributes to the resistant phenotype, we observed that the rate of efflux of dansylated estramustine was increased in SKEM compared with control cells. In addition, antisense treatment directed toward ABC2 mRNA sensitized the resistant cells to estramustine. Together, these results suggest that amplification and overexpression of ABC2 contributes to estramustine resistance and provides the first indication of a potential cellular function for this product.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/physiology , Antineoplastic Agents, Hormonal/pharmacokinetics , Carcinoma/genetics , Carcinoma/metabolism , Estramustine/pharmacokinetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , ATP-Binding Cassette Transporters/biosynthesis , Antineoplastic Agents, Hormonal/pharmacology , Carcinoma/pathology , Chromosomes, Human, Pair 9 , Down-Regulation , Drug Resistance, Neoplasm , Estramustine/pharmacology , Female , Gene Amplification , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Oligonucleotides, Antisense/pharmacology , Ovarian Neoplasms/pathology , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic , Tumor Cells, Cultured
16.
Biochem Pharmacol ; 54(6): 649-55, 1997 Sep 15.
Article in English | MEDLINE | ID: mdl-9310341

ABSTRACT

L1210 MQ-580 is a murine leukemia cell line resistant to the cytotoxic activity of the alpha-(N)-heterocyclic carboxaldehyde thiosemicarbazone class of inhibitors of ribonucleotide reductase. The line is cross-resistant to etoposide, daunomycin, and vinblastine. L1210 MQ-580 cells expressed 8-fold resistance to 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP), a relatively newly developed inhibitor of ribonucleotide reductase. The accumulation of [14C]3-AP by L1210 MQ-580 cells was 5- to 6-fold less than by parental L1210 cells. An increased rate of efflux of 3-AP was responsible for the lower steady-state concentration of 3-AP in resistant cells. In reverse transcription-polymerase chain reaction assays, L1210 MQ-580 cells were found to overexpress the multidrug resistance genes mdr1, mdr3, and mrp, but not the mdr2 gene, compared with parental L1210 cells. Measurement of the steady-state concentration of doxorubicin, a potential substrate for both the mdr and mrp gene products, demonstrated that L1210 MQ-580 cells accumulated 4-fold less anthracycline than parental cells. These findings indicate that drug efflux is a major determinant of the pattern of cross-resistance of L1210 MQ-580 cells. To extrapolate these observations to the human homologues of the mdr1, mdr3, and mrp murine genes, the effects of 3-AP were measured in L1210/VMDRC0.06 and NIH3T3 36-8-32 cells transfected with human MDR1 and MRP cDNAs, respectively. The transfectants were 2- to 3-fold resistant to the cytotoxic effects of 3-AP and accumulated less [14C]3-AP than their parental mock-transfected counterparts. Moreover, the cytotoxic activity of 3-AP was significantly greater in two double mrp gene knockout cell lines than in parental W 9.5 embryonic stem cells. Thus, the results suggest that 3-AP is a substrate for both the P-glycoprotein and MRP and that baseline MRP expression has the capacity to exert a protective role against the toxicity of this agent.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B , ATP-Binding Cassette Transporters/genetics , Enzyme Inhibitors/pharmacology , Leukemia L1210/genetics , Ribonucleotide Reductases/antagonists & inhibitors , Animals , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Humans , Leukemia L1210/pathology , Mice , Multidrug Resistance-Associated Proteins , Thiosemicarbazones/pharmacology
17.
J Biol Chem ; 272(30): 18905-9, 1997 Jul 25.
Article in English | MEDLINE | ID: mdl-9228069

ABSTRACT

The largest subunit of RNA polymerase II contains a C-terminal repeated domain (CTD) that is the site of phosphorylation by serine (threonine) and tyrosine kinases. Phosphorylation of the CTD is correlated with transcription elongation. A number of different kinases have previously been shown to phosphorylate the CTD; among them is a nuclear tyrosine kinase encoded by the c-abl proto-oncogene. The processive and high stoichiometric phosphorylation of RNA polymerase II by c-Abl requires the tyrosine kinase, the SH2 domain, and a CTD-interacting domain (CTD-ID) in the Abl protein. The physiological tyrosine phosphorylation of RNA polymerase II by c-Abl in DNA damage response has previously been demonstrated. Basal tyrosine phosphorylation of RNA polymerase II, however, is observed in cells derived from abl-deficient mice, indicating the existence of other CTD tyrosine kinases. In this report, we show that the tyrosine kinase encoded by an Abl-related gene (Arg) also phosphorylates the CTD in vitro and in transfected cells. The SH2 and kinase domain of Arg are 95% identical to that of c-Abl. However, these two proteins share only 29% identity in the large C-terminal region. Interestingly, a CTD-ID is also found in the C-terminal region of Arg. Mapping studies and sequence analysis have led to the identification of the CTD-ID that is highly conserved among the divergent C-terminal regions of Abl and Arg. These results indicate that tyrosine phosphorylation of RNA polymerase II CTD could be catalyzed by either c-Abl or Arg kinase.


Subject(s)
Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-abl/metabolism , RNA Polymerase II/metabolism , Tyrosine/metabolism , Amino Acid Sequence , Animals , Binding Sites , COS Cells , CSK Tyrosine-Protein Kinase , Conserved Sequence , Humans , Mice , Molecular Sequence Data , Peptide Fragments/metabolism , Phosphorylation , Protein Kinases/metabolism , Protein-Tyrosine Kinases/chemistry , Proto-Oncogene Mas , src Homology Domains , src-Family Kinases
18.
J Biol Chem ; 272(28): 17542-50, 1997 Jul 11.
Article in English | MEDLINE | ID: mdl-9211900

ABSTRACT

Arg and c-Abl represent the mammalian members of the Abelson family of protein-tyrosine kinases. A novel Arg/Abl-binding protein, ArgBP2, was isolated using a segment of the Arg COOH-terminal domain as bait in the yeast two-hybrid system. ArgBP2 contains three COOH-terminal Src homology 3 domains, a serine/threonine-rich domain, and several potential Abl phosphorylation sites. ArgBP2 associates with and is a substrate of Arg and v-Abl, and is phosphorylated on tyrosine in v-Abl-transformed cells. ArgBP2 is widely expressed in human tissues and extremely abundant in heart. In epithelial cells ArgBP2 is located in stress fibers and the nucleus, similar to the reported localization of c-Abl. In cardiac muscle cells ArgBP2 is located in the Z-disks of sarcomeres. These observations suggest that ArgBP2 functions as an adapter protein to assemble signaling complexes in stress fibers, and that ArgBP2 is a potential link between Abl family kinases and the actin cytoskeleton. In addition, the localization of ArgBP2 to Z-disks suggests that ArgBP2 may influence the contractile or elastic properties of cardiac sarcomeres and that the Z-disk is a target of signal transduction cascades.


Subject(s)
Homeodomain Proteins/metabolism , Myocardium/metabolism , Oncogene Proteins v-abl/metabolism , src Homology Domains , 3T3 Cells , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Arginine/metabolism , COS Cells , Chickens , Fluorescent Antibody Technique, Indirect , Homeodomain Proteins/chemistry , Humans , Mice , Molecular Sequence Data , Myocardial Contraction , Phosphorylation , Proline/metabolism , RNA-Binding Proteins , Sarcomeres/metabolism , Spodoptera , Substrate Specificity , Tissue Distribution , Transfection
19.
Oncogene ; 14(23): 2793-801, 1997 Jun 12.
Article in English | MEDLINE | ID: mdl-9190895

ABSTRACT

The AKT2 oncogene encodes a protein-serine/threonine kinase containing a pleckstrin homology domain characteristic of many signaling proteins. Recently, it was shown that AKT2 kinase activity can be induced by platelet-derived growth factor through phosphatidylinositol-3-OH kinase, suggesting that AKT2 may be an important signal mediator that contributes to the control of cell proliferation. We previously reported amplification and overexpression of AKT2 in human cancers. To investigate the transforming activity of AKT2, we used a retrovirus-based construct to express AKT2 in NIH3T3 cells. Overexpression of AKT2 was found to transform NIH3T3 cells, as determined by growth in soft agar and tumor formation in nude mice. The oncogenic activity of AKT2 was diminished by truncation of a 70-amino acid proline-rich region at the carboxyl-terminus. To facilitate the characterization of AKT2, we generated monoclonal and polyclonal antibodies against this protein. AKT2 was localized to the cytoplasm by cell fractionation experiments, immunocytochemistry, and immunofluorescence. Protein levels were more abundant in mitotic cells than in interphase cells. Western blot analysis of synchronized pancreatic cancer cells demonstrated that the expression level of AKT2 protein in mitotic cells is three to fivefold higher than in their interphase counterparts. A time-course study of phytohemagglutinin-stimulated lymphocytes revealed that AKT2 mRNA and AKT2 protein levels are highest 48-72 h after addition of mitogen, when cells are actively dividing. These data suggest that AKT2 could play a significant role in cell cycle progression and that the oncogenic activity of overexpressed AKT2 may be mediated by aberrant regulation of cellular proliferation.


Subject(s)
Cell Cycle/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Oncogene Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins , 3T3 Cells , Animals , Antibodies, Monoclonal , Fibroblasts/metabolism , Fluorescent Antibody Technique, Indirect , Humans , Interphase , Mice , Mice, Nude , Mitosis , Oncogene Proteins/immunology , Oncogene Proteins/metabolism , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt , Subcellular Fractions/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...