ABCC8

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 11967 Experts worldwide ranked by ideXlab platform

Jeremy A. Hirota - One of the best experts on this subject based on the ideXlab platform.

  • interactions between abcc4 mrp4 and abcc7 cftr in human airway epithelial cells in lung health and disease
    The International Journal of Biochemistry & Cell Biology, 2021
    Co-Authors: Jenny P Nguyen, Yechan Kim, Quynh T Cao, Jeremy A. Hirota
    Abstract:

    ATP binding cassette (ABC) transporters are present in all three domains of life - Archaea, Bacteria, and Eukarya. The conserved nature is a testament to the importance of these transporters in regulating endogenous and exogenous substrates required for life to exist. In humans, 49 ABC transporters have been identified to date with broad expression in different lung cell types with multiple transporter family members contributing to lung health and disease. The ABC transporter most commonly known to be linked to lung pathology is ABCC7, also known as cystic fibrosis transmembrane conductance regulator - CFTR. Closely related to the CFTR genomic sequence is ABCC4/multi-drug resistance protein-4. Genomic proximity is shared with physical proximity, with ABCC4 and CFTR physically coupled in cell membrane microenvironments of epithelial cells to orchestrate functional consequences of cyclic-adenosine monophosphate (cAMP)-dependent second messenger signaling and extracellular transport of endogenous and exogenous substrates. The present concise review summarizes the emerging data defining a role of the (ABCC7/CFTR)-ABCC4 macromolecular complex in human airway epithelial cells as a physiologically important pathway capable of impacting endogenous and exogenous mediator transport and ion transport in both lung health and disease.

  • the impact of cigarette smoke exposure copd or asthma status on abc transporter gene expression in human airway epithelial cells
    Scientific Reports, 2019
    Co-Authors: Jennifer A Aguiar, Andrea Tamminga, Ryan D Huff, Jenny P Nguyen, Anna Dvorkingheva, Briallen Lobb, Martin R Stampfli, Andrew C. Doxey, Jeremy A. Hirota
    Abstract:

    ABC transporters are conserved in prokaryotes and eukaryotes, with humans expressing 48 transporters divided into 7 classes (ABCA, ABCB, ABCC, ABCD, ABDE, ABCF, and ABCG). Throughout the human body, ABC transporters regulate cAMP levels, chloride secretion, lipid transport, and anti-oxidant responses. We used a bioinformatic approach complemented with in vitro experimental methods for validation of the 48 known human ABC transporters in airway epithelial cells using bronchial epithelial cell gene expression datasets available in NCBI GEO from well-characterized patient populations of healthy subjects and individuals that smoke cigarettes, or have been diagnosed with COPD or asthma, with validation performed in Calu-3 airway epithelial cells. Gene expression data demonstrate that ABC transporters are variably expressed in epithelial cells from different airway generations, regulated by cigarette smoke exposure (ABCA13, ABCB6, ABCC1, and ABCC3), and differentially expressed in individuals with COPD and asthma (ABCA13, ABCC1, ABCC2, ABCC9). An in vitro cell culture model of cigarette smoke exposure was able to recapitulate select observed in situ changes. Our work highlights select ABC transporter candidates of interest and a relevant in vitro model that will enable a deeper understanding of the contribution of ABC transporters in the respiratory mucosa in lung health and disease.

Heyo K. Kroemer - One of the best experts on this subject based on the ideXlab platform.

  • Role of the ABC-transporters ABCC4, ABCC5 and ABCC11 in response to chemotherapy in adult patients with acute myeloid leukemia
    Cancer Research, 2005
    Co-Authors: Christoph A. Ritter, Kathleen Kock, Gabriele Jedlitschky, James F. Beck, Gerhard Ehninger, Heyo K. Kroemer, Markus Schaich
    Abstract:

    6169 Resistance to chemotherapeutic drugs is a major factor limiting a successful therapy. One proposed mechanism for treatment failure in patients with acute myeloid leukemia (AML) is the efflux of cytostatics. The multidrug resistance-associated proteins ABCC4, ABCC5 and ABCC11 have the ability to transport cyclic nucleotides and anti-cancer nucleoside analogs. These substances are particularly valuable for the treatment of AML, which is the most common form of leukemia in adults. Therefore we examined the clinical relevance of ABCC4, ABCC5 and ABCC11 by analyzing their mRNA expression levels in pre-treatment leukemic blast cells of the bone marrow of 50 newly diagnosed AML-patients participating in the SHG-AML-96 study. mRNA expression levels were investigated by quantitative TaqMan Real-time PCR and normalized to 18S mRNA expression. The median relative mRNA expression levels of the patient probes were different between the three examined genes (28.8 (ABCC4), 63.7 (ABCC5), 0.34 (ABCC11)). There was no significant association with age, white blood cell count, frequency of blast cells in bone marrow and P-gp mRNA level, but patients with the French-American-British (FAB) subtype M1 showed a higher mRNA expression of all genes examined (P ABCC4 =0.002; P ABCC5 =0.007; P ABCC11 =0.004). For ABCC4 and ABCC5 no significant association was found for complete remission rate and overall survival. However, Kaplan-Meyer statistics and Log-rank test suggest that despite its low mRNA expression ABCC11 has a negative influence on overall survival with a 5 year probability of survival of 28% for patients with low and only 8% for patients with high ABCC11 expression (P=0.03). As complete remission rate was not affected this was mainly due to a higher relapse rate in high ABCC11 expressing patients. In conclusion, ABCC11 expression seems to be a new promising predictor for treatment outcome in adult AML.

  • Cellular export of drugs and signaling molecules by the ATP-binding cassette transporters MRP4 (ABCC4) and MRP5 (ABCC5).
    Drug Metabolism Reviews, 2005
    Co-Authors: Christoph A. Ritter, Kathleen Kock, Gabriele Jedlitschky, Heyo K. Kroemer, Henriette E. Meyer Zu Schwabedissen, Markus Grube
    Abstract:

    Like other members of the multidrug resistance protein (MRP)/ABCC subfamily of ATP-binding cassette transporters, MRP4 (ABCC4) and MRP5 (ABCC5) are organic anion transporters. They have, however, the outstanding ability to transport nucleotides and nucleotide analogs. In vitro experiments using drug-selected or -transfected cells indicated that these transport proteins, when overexpressed, can lower the intracellular concentration of nucleoside/nucleotide analogs, such as the antiviral compounds PMEA (9-(2-phosphonylmethoxyethyl)adenine) or ganciclovir, and of anticancer nucleobase analogs, such as 6-mercaptopurine, after their conversion into the respective nucleotides. This may lead to an impaired ability of these compounds to inhibit virus replication or cell proliferation. It remains to be tested whether antiviral or anticancer chemotherapy based on nucleobase, nucleoside, or nucleotide precursors can be modulated by inhibition of MRP4 and MRP5. MRP4 also seems to be able to mediate the transport of conjugated steroids, prostaglandins, and glutathione. Furthermore, cyclic nucleotides (cyclic adenosine monophosphate and cyclic guanine monophosphate) are exported from cells by MRP4 and MRP5. This may modulate the intracellular concentration of these important mediators, besides the action of phosphodiesterases, as well as provide extracellular nucleotides for a possible paracrine action. In this line, tissue distribution and subcellular localization of MRP4 and MRP5 specifically in smooth muscle cells (MRP5), platelet-dense granules (MRP4), and nervous cells (MRP4 and MRP5), besides the capillary endothelium, point not only to a possible function of these transporters as exporters in cellular defense, but also to a physiological function in signaling processes.

Theresa M C Tan - One of the best experts on this subject based on the ideXlab platform.

  • multidrug resistance protein 4 mrp4 abcc4 mediates efflux of bimane glutathione
    The International Journal of Biochemistry & Cell Biology, 2004
    Co-Authors: Jing Bai, Liqi Lai, Hui Ching Yeo, Boon Cher Goh, Theresa M C Tan
    Abstract:

    Abstract Multidrug resistance proteins (MRPs) are ATP-dependent export pumps that mediate the export of organic anions. ABCC1 (MRP1), ABCC2 (MRP2) and ABCC3 (MRP3) are all able to facilitate the efflux of anionic conjugates including glutathione (GSH), glucuronide and sulfate conjugates of xenobiotics and endogenous molecules. Earlier studies showed that ABCC4 functions as an ATP-driven export pump for cyclic AMP and cyclic GMP, as well as estradiol-17-β- d -glucuronide. However, it was unclear if other conjugated metabolites can be transported by ABCC4. Hence in this study, a fluorescent substrate, bimane-glutathione (bimane-GS) was used to further examine the transport activity of ABCC4. Using cells stably overexpressing ABCC4, this study shows that ABCC4 can facilitate the efflux of the glutathione conjugate, bimane-glutathione. Bimane-glutathione efflux increased with time and >85% of the conjugate was exported after 15 min. This transport was abolished in the presence of 2.5 μM carbonylcyanide m-chlorophenylhydrasone (CCCP), an uncoupler of oxidative phosphorylation. Inhibition was also observed with known inhibitors of MRP transporters including benzbromarone, verapamil and indomethacin. In addition, 100 μM methotrexate, an ABCC4 substrate or 100 μM 6-thioguanine (6-TG), a compound whose monophosphate metabolite is an ABCC4 substrate, reduced efflux by >40%. A concentration-dependent inhibition of bimane-glutathione efflux was observed with 1-chloro-2,4-dinitrobenzene (CDNB) which is metabolized intracellularly to the glutathione conjugate, 2,4-dinitrophenyl-glutathione (DNP-GS). The determination that ABCC4 can mediate the transport of glucuronide and glutathione conjugates indicates that ABCC4 may play a role in the cellular extrusion of Phase II detoxification metabolites.

  • role of glutathione in the multidrug resistance protein 4 mrp4 abcc4 mediated efflux of camp and resistance to purine analogues
    Biochemical Journal, 2002
    Co-Authors: Liqi Lai, Theresa M C Tan
    Abstract:

    Multidrug resistance protein 4 (MRP4/ABCC4) is a member of the MRP subfamily, which in turn is a member of the superfamily of ATP-binding-cassette (ABC) transporters. Within the MRP subfamily, ABCC4,ABCC5 (MRP5), ABCC11 (MRP8) and ABCC12 (MRP9) have similar predicted membrane topologies. All lack the additional transmembrane domain, TMD(0), which is present in the other MRPs. Using cells stably overexpressing ABCC4, this study shows that ABCC4 exports GSH. ABCC4 also facilitates the efflux of cAMP. Depletion of intracellular GSH with DL-buthionine-(S,R)-sulphoximine led to decreased export of cAMP and a corresponding increase in intracellular cAMP was observed. ABCC4 also mediates resistance to purine analogues 9-(2-phosphonylmethoxyethyl)-adenine and 6-thioguanine. This resistance can be reversed by the presence of DL-buthionine-(S,R)-sulphoximine. We conclude that as well as nucleotide and nucleoside analogues, ABCC4 can mediate the export of GSH. In addition, GSH plays an important role in the function of ABCC4. Depletion of intracellular GSH adversely affects the export of cAMP by ABCC4. Resistance to nucleoside analogues is also adversely affected by depletion of cellular GSH.

Dietrich Keppler - One of the best experts on this subject based on the ideXlab platform.

  • atp dependent transport of leukotrienes b4 and c4 by the multidrug resistance protein abcc4 mrp4
    Journal of Pharmacology and Experimental Therapeutics, 2008
    Co-Authors: Maria Rius, Johanna Hummeleisenbeiss, Dietrich Keppler
    Abstract:

    The proinflammatory mediators leukotriene (LT) B(4) and LTC(4) must be transported out of cells before they can interact with LT receptors. Previously, we identified the multidrug resistance protein ABCC1 (MRP1) as an efflux pump for LTC(4). However, the molecular basis for the efflux of LTB(4) was unknown. Here, we demonstrate that human ABCC4 mediates the ATP-dependent efflux of LTB(4) in the presence of reduced glutathione (GSH), whereby the latter can be replaced by S-methyl GSH. Transport studies were performed with inside-out membrane vesicles from V79 fibroblasts and Sf9 insect cells that contained recombinant ABCC4, with vesicles from human platelets and myelomonocytic U937 cells, which were rich in endogenous ABCC4, but ABCC1 was below detectability. Moreover, human polymorphonuclear leukocytes contained ABCC4. K(m) values for LTB(4) were 5.2 muM with vesicles from fibroblasts and 5.6 muM with vesicles from platelets. ABCC4, with its broad substrate specificity, also functioned as an ATP-dependent efflux pump for LTC(4) with a K(m) of 0.13 muM in vesicles from fibroblasts and 0.32 muM in vesicles from platelets. However, GSH was not required for the transport of this glutathionylated leukotriene. The transport of LTC(4) by ABCC4 explains its release from platelets during transcellular synthesis. ATP-dependent transport of LTB(4) and LTC(4) by ABCC4 was inhibited by several organic anions, including S-decyl GSH, sulindac sulfide, and by the LTD(4) receptor antagonists montelukast and 3-(((3-(2-(7-chloro-2-quinolinyl)ethenyl)phenyl)-((3-dimethyl-amino-3-oxopropyl)-thio)-methyl)thio)propanoic acid (MK571). Thus, as an efflux pump for the proinflammatory mediators LTB(4) and LTC(4), ABCC4 may represent a novel target for anti-inflammatory therapies.

  • The apical conjugate efflux pump ABCC2 (MRP2)
    Pflugers Archiv European Journal of Physiology, 2007
    Co-Authors: Anne T Nies, Dietrich Keppler
    Abstract:

    ABCC2 is a member of the multidrug resistance protein subfamily localized exclusively to the apical membrane domain of polarized cells, such as hepatocytes, renal proximal tubule epithelia, and intestinal epithelia. This localization supports the function of ABCC2 in the terminal excretion and detoxification of endogenous and xenobiotic organic anions, particularly in the unidirectional efflux of substances conjugated with glutathione, glucuronate, or sulfate, as exemplified by leukotriene C(4), bilirubin glucuronosides, and some steroid sulfates. The hepatic ABCC2 pump contributes to the driving forces of bile flow. Acquired or hereditary deficiency of ABCC2, the latter known as Dubin-Johnson syndrome in humans, causes an increased concentration of bilirubin glucuronosides in blood because of their efflux from hepatocytes via the basolateral ABCC3, which compensates for the deficiency in ABCC2-mediated apical efflux. In this article we provide an overview on the molecular characteristics of ABCC2 and its expression in various tissues and species. We discuss the transcriptional and posttranscriptional regulation of ABCC2 and review approaches to the functional analysis providing information on its substrate specificity. A comprehensive list of sequence variants in the human ABCC2 gene summarizes predicted and proven functional consequences, including variants leading to Dubin-Johnson syndrome.

  • ABCC Drug Efflux Pumps and Organic Anion Uptake Transporters in Human Gliomas and the Blood-Tumor Barrier
    Cancer Research, 2005
    Co-Authors: Holger Bronger, Dietrich Keppler, Jörg König, Kathrin Kopplow, Hans-herbert Steiner, Rezvan Ahmadi, Christel Herold-mende, Anne T Nies
    Abstract:

    Delivery of therapeutic agents to the brain and its neoplasms depends on the presence of membrane transport proteins in the blood-brain barrier and in the target cells. The cellular and subcellular localization of these membrane transporters determines the drug accessibility to the brain and its tumors. We therefore analyzed the expression and localization of six members of the multidrug resistance protein family of ATP-dependent efflux pumps (ABCC1-ABCC6, formerly MRP1-MRP6) and of six organic anion uptake transporters (OATP1A2, OATP1B1, OATP1B3, OATP1C1, OATP2B1, and OATP4A1) in 61 human glioma specimens of different histologic subtypes. Real-time PCRs indicated expressions of ABCC1, ABCC3, ABCC4, and ABCC5. In addition, we detected expressions of the OATP uptake transporter genes SLCO1A2, SLCO1C1, SLCO2B1, and SLCO4A1. At the protein level, however, only OATP1A2 and OATP2B1 were detectable by immunofluorescence microscopy in the luminal membrane of endothelial cells forming the blood-brain barrier and the blood-tumor barrier, but not in the glioma cells. ABCC4 and ABCC5 proteins were the major ABCC subfamily members in gliomas, localized both at the luminal side of the endothelial cells and in the glioma cells of astrocytic tumors and in the astrocytic portions of oligoastrocytomas. These results indicate that expression of ABCC4 and ABCC5 is associated with an astrocytic phenotype, in accordance with their expression in astrocytes and with the higher chemoresistance of astrocytic tumors as compared with oligodendrogliomas. Our data provide a basis for the assessment of the role of uptake transporters and efflux pumps in the accessibility of human gliomas for chemotherapeutic agents.

Susan P C Cole - One of the best experts on this subject based on the ideXlab platform.

  • Mutagenic Analysis of the Putative ABCC6 Substrate-Binding Cavity Using a New Homology Model
    'MDPI AG', 2021
    Co-Authors: Flora Szeri, Susan P C Cole, Gwenaelle Conseil, Valentina Corradi, Fatemeh Niaziorimi, Sylvia Donnelly, Peter D. Tieleman, Koen Van De Wetering
    Abstract:

    Inactivating mutations in ABCC6 underlie the rare hereditary mineralization disorder pseudoxanthoma elasticum. ABCC6 is an ATP-binding cassette (ABC) integral membrane protein that mediates the release of ATP from hepatocytes into the bloodstream. The released ATP is extracellularly converted into pyrophosphate, a key mineralization inhibitor. Although ABCC6 is firmly linked to cellular ATP release, the molecular details of ABCC6-mediated ATP release remain elusive. Most of the currently available data support the hypothesis that ABCC6 is an ATP-dependent ATP efflux pump, an un-precedented function for an ABC transporter. This hypothesis implies the presence of an ATP-binding site in the substrate-binding cavity of ABCC6. We performed an extensive mutagenesis study using a new homology model based on recently published structures of its close homolog, bovine Abcc1, to characterize the substrate-binding cavity of ABCC6. Leukotriene C4 (LTC4), is a high-affinity substrate of ABCC1. We mutagenized fourteen amino acid residues in the rat ortholog of ABCC6, rAbcc6, that corresponded to the residues in ABCC1 found in the LTC4 binding cavity. Our functional characterization revealed that most of the amino acids in rAbcc6 corresponding to those found in the LTC4 binding pocket in bovine Abcc1 are not critical for ATP efflux. We conclude that the putative ATP binding site in the substrate-binding cavity of ABCC6/rAbcc6 is distinct from the bovine Abcc1 LTC4-binding site

  • cysteinyl leukotriene receptor 1 2 antagonists nonselectively modulate organic anion transport by multidrug resistance proteins mrp1 4
    Drug Metabolism and Disposition, 2016
    Co-Authors: Mark Csandl, Gwenaelle Conseil, Susan P C Cole
    Abstract:

    Active efflux of both drugs and organic anion metabolites is mediated by the multidrug resistance proteins (MRPs). MRP1 (ABCC1), MRP2 (ABCC2), MRP3 (ABCC3), and MRP4 (ABCC4) have partially overlapping substrate specificities and all transport 17β-estradiol 17-(β-d-glucuronide) (E217βG). The cysteinyl leukotriene receptor 1 (CysLT1R) antagonist MK-571 inhibits all four MRP homologs, but little is known about the modulatory effects of newer leukotriene modifiers (LTMs). Here we examined the effects of seven CysLT1R- and CysLT2R-selective LTMs on E217βG uptake into MRP1-4-enriched inside-out membrane vesicles. Their effects on uptake of an additional physiologic solute were also measured for MRP1 [leukotriene C4 (LTC4)] and MRP4 [prostaglandin E2 (PGE2)]. The two CysLT2R-selective LTMs studied were generally more potent inhibitors than CysLT1R-selective LTMs, but neither class of antagonists showed any MRP selectivity. For E217βG uptake, LTM IC50s ranged from 1.2 to 26.9 μM and were most comparable for MRP1 and MRP4. The LTM rank order inhibitory potencies for E217βG versus LTC4 uptake by MRP1, and E217βG versus PGE2 uptake by MRP4, were also similar. Three of four CysLT1R-selective LTMs also stimulated MRP2 (but not MRP3) transport and thus exerted a concentration-dependent biphasic effect on MRP2. The fourth CysLT1R antagonist, LY171883, only stimulated MRP2 (and MRP3) transport but none of the MRPs were stimulated by either CysLT2R-selective LTM. We conclude that, in contrast to their CysLTR selectivity, CysLTR antagonists show no MRP homolog selectivity, and data should be interpreted cautiously if obtained from LTMs in systems in which more than one MRP is present.

  • transmembrane transport of endo and xenobiotics by mammalian atp binding cassette multidrug resistance proteins
    Physical Review, 2006
    Co-Authors: Roger G Deeley, Christopher J Westlake, Susan P C Cole
    Abstract:

    Multidrug Resistance Proteins (MRPs), together with the cystic fibrosis conductance regulator (CFTR/ABCC7) and the sulfonylurea receptors (SUR1/ABCC8 and SUR2/ABCC9) comprise the 13 members of the ...

  • polymorphisms of mrp1 abcc1 and related atp dependent drug transporters
    Pharmacogenetics and Genomics, 2005
    Co-Authors: Gwenaelle Conseil, Roger G Deeley, Susan P C Cole
    Abstract:

    Genetic variations in drug metabolizing enzymes and targets are established determinants of adverse drug reactions and interactions, but less is known about the role of genetic polymorphisms in membrane transport proteins. MRP1 (ABCC1) is one of 13 polytopic membrane proteins that comprise the 'C' subfamily of the ATP-binding cassette (ABC) superfamily of transport proteins. MRP1 and related ABCC family members, including MRP2, 3, 4 and 5 (ABCC2, 3, 4 and 5), each have a distinctive pattern of tissue expression and substrate specificity. Together, these five transporters play important roles in the disposition and elimination of drugs and other organic anions, and in maintenance of blood-tissue barriers, as confirmed by enhanced chemosensitivity of respective knockout mice. Moreover, Mrp2 (Abcc2) deficient animals display mild conjugated hyperbilirubinemia, corresponding to a human condition known as Dubin-Johnson syndrome (DJS). Naturally occurring mutations in MRP/ABCC-related drug transporters have been reported, some of which are non-synonymous single nucleotide polymorphisms. The consequences of the resulting amino acid changes can sometimes be predicted from in vitro site-directed mutagenesis studies or from knowledge of mutations of analogous (conserved) residues in ABCC proteins that cause DJS, Pseudoxanthoma elasticum (ABCC6), cystic fibrosis (CFTR/ABCC7) or persistent hyperinsulinemic hypoglycemia of infancy (SUR1/ABCC8). Continual updating of databases of sequence variants and haplotype analysis, together with in vitro biochemical validation assays and pharmacological studies in knockout animals, should make it possible to determine how genetic variation in the MRP-related transporters contributes to the range of responses to drugs and chemicals observed in different human populations.