Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Adv Sci (Weinh) ; 11(6): e2307271, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38072640

ABSTRACT

Chemotherapy is widely used to treat colorectal cancer (CRC). Despite its substantial benefits, the development of drug resistance and adverse effects remain challenging. This study aimed to elucidate a novel role of glucagon in anti-cancer therapy. In a series of in vitro experiments, glucagon inhibited cell migration and tube formation in both endothelial and tumor cells. In vivo studies demonstrated decreased tumor blood vessels and fewer pseudo-vessels in mice treated with glucagon. The combination of glucagon and chemotherapy exhibited enhanced tumor inhibition. Mechanistic studies demonstrated that glucagon increased the permeability of blood vessels, leading to a pronounced disruption of vessel morphology. Signaling pathway analysis identified a VEGF/VEGFR-dependent mechanism whereby glucagon attenuated angiogenesis through its receptor. Clinical data analysis revealed a positive correlation between elevated glucagon expression and chemotherapy response. This is the first study to reveal a role for glucagon in inhibiting angiogenesis and vascular mimicry. Additionally, the delivery of glucagon-encapsulated PEGylated liposomes to tumor-bearing mice amplified the inhibition of angiogenesis and vascular mimicry, consequently reinforcing chemotherapy efficacy. Collectively, the findings demonstrate the role of glucagon in inhibiting tumor vessel network and suggest the potential utility of glucagon as a promising predictive marker for patients with CRC receiving chemotherapy.


Subject(s)
Colorectal Neoplasms , Glucagon , Humans , Animals , Mice , Glucagon/pharmacology , Glucagon/therapeutic use , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Colorectal Neoplasms/pathology , Signal Transduction , Cell Line, Tumor
2.
Sci Rep ; 13(1): 13956, 2023 08 25.
Article in English | MEDLINE | ID: mdl-37626095

ABSTRACT

Corneal keratoconus (KC) is a dilated (ectatic) corneal disease characterized by a central thinning of the cornea, which causes protrusion into a conical shape that seriously affects vision. However, due to the complex etiology of keratoconus, its entire mechanism remains unclear and there is no mechanism-directed treatment method. Ferroptosis is a novel programmed cell death mechanism related to lipid peroxidation, stress, and amino acid metabolism, which plays a crucial role in various diseases. This study aimed to explore the relationship between keratoconus and ferroptosis, to provide new insights into the mechanism of keratoconus development, and potential treatment options based on further elucidation of this mechanism. The corresponding mRNA microarray expression matrix data of KC patients were obtained from GEO database (GSE204791). Weighted co-expression network analysis (WGCNA) and support vector machine recursive feature elimination (SVM-RFE) were selected to screen hub genes, which were overlapped with ferroptosis genes (FRGs) from FerrDb. GO and GSEA were performed to analyze differential pathways, ssGSEA was used to determine immune status, and then, feasible drugs were predicted by gene-drug network. Additionally, we predicted the miRNA and IncRNA of hub genes to identify the underlying mechanism of disease so as to predict treatment for the disease. The epithelial transcriptome from keratoconus tissue mRNA microarray data (GSE204791) was extracted for the main analysis, including eight epithelial cells and eight epithelial control cells. The differential genes that were overlapped by WGCAN, SVM-RFE and FRGs were mainly related to oxidative stress, immune regulation, cellular inflammation, and metal ion transport. Through further analysis, aldo-keto reductase family 1 member C3 (AKR1C3) was selected, and negatively correlated with mature CD56 natural killer (NK) cells and macrophages. Then, gene-drug interaction network analysis and miRNA prediction were performed through the website. It was concluded that four immune-related drugs (INDOMETHACIN, DAUNORUBICIN, DOXORUBICIN, DOCETAXEL) and a miRNA (has-miR-184) were screened to predict potential drugs and targets for disease treatment. To our knowledge, this was the first report of KC being associated with ferroptosis and prompted search for differential genes to predict drug targets of gene immunotherapy. Our findings provided insight and a solid basis for the analysis and treatment of KC.


Subject(s)
Ferroptosis , Keratoconus , MicroRNAs , Humans , Keratoconus/genetics , Ferroptosis/genetics , MicroRNAs/genetics , Cornea , Apoptosis
3.
J Nanobiotechnology ; 21(1): 9, 2023 Jan 06.
Article in English | MEDLINE | ID: mdl-36609374

ABSTRACT

As one of the most significant imaging modalities currently available, magnetic resonance imaging (MRI) has been extensively utilized for clinically accurate cancer diagnosis. However, low signal-to-noise ratio (SNR) and low specificity for tumors continue to pose significant challenges. Inspired by the distance-dependent magnetic resonance tuning (MRET) phenomenon, the tumor microenvironment (TME)-activated off-on T1-T2 dual-mode MRI nanoswitch is presented in the current study to realize the sensitive early diagnosis of tumors. The tumor-specific nanoswitch is designed and manufactured on the basis of PDGFB-conjugating ferroferric oxide coated by Mn-doped silica (PDGFB-FMS), which can be degraded under the high-concentration GSH and low pH in TME to activate the T1-T2 dual-mode MRI signals. The tumor-specific off-on dual-mode MRI nanoswitch can significantly improve the SNR and is used successfully for the accurate diagnosis of early-stage tumors, particularly for orthotopic prostate cancer. In addition, the systemic delivery of the nanoswitch did not cause blood or tissue damage, and it can be excreted out of the body in a timely manner, demonstrating excellent biosafety. Overall, the strategy is a significant step in the direction of designing off-on dual-mode MRI nanoprobes to improve imaging accuracy, which opens up new avenues for the development of new MRI probes.


Subject(s)
Contrast Media , Neoplasms , Male , Humans , Proto-Oncogene Proteins c-sis , Neoplasms/diagnostic imaging , Magnetic Resonance Imaging/methods , Silicon Dioxide , Tumor Microenvironment
4.
J Biol Chem ; 288(31): 22207-18, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23766510

ABSTRACT

The ABC transporter ABCC4 is recognized as an ATP-dependent exporter of endogenous substances as well as an increasing variety of anionic chemotherapeutics. A loss-of-function variant of zebrafish Abcc4 was identified with a single amino acid substitution in the cytoplasmic loop T804M. Because this substituted amino acid is highly conserved among ABCC4 orthologs and is located in cytoplasmic loop 3 (CL3), we investigated the impact of this mutation on human and zebrafish Abcc4 expression. We demonstrate that zebrafish Abcc4 T804M or human ABCC4 T796M exhibit substantially reduced expression, coupled with impaired plasma membrane localization. To understand the molecular basis for the localization defect, we developed a homology model of zebrafish Abcc4. The homology model suggested that the bulky methionine substitution disrupted side-chain contacts. Molecular dynamic simulations of a fragment of human or zebrafish CL3 containing a methionine substitution indicated altered helicity coupled with reduced thermal stability. Trifluoroethanol challenge coupled with circular dichroism revealed that the methionine substitution disrupted the ability of this fragment of CL3 to readily form an α-helix. Furthermore, expression and plasma membrane localization of these mutant ABCC4/Abcc4 proteins are mostly rescued by growing cells at subphysiological temperatures. Because the cystic fibrosis transmembrane conductance regulator (ABCC7) is closely related to ABCC4, we extended this by engineering certain pathogenic CFTR-CL3 mutations, and we showed they destabilized human and zebrafish ABCC4. Altogether, our studies provide the first evidence for a conserved domain in CL3 of ABCC4 that is crucial in ensuring its proper plasma membrane localization.


Subject(s)
Cytoplasm/metabolism , Multidrug Resistance-Associated Proteins/genetics , Phylogeny , Amino Acid Sequence , Animals , Cell Line , Humans , Mice , Models, Molecular , Molecular Sequence Data , Multidrug Resistance-Associated Proteins/chemistry , Multidrug Resistance-Associated Proteins/metabolism , NIH 3T3 Cells , Sequence Homology, Amino Acid , Zebrafish
5.
J Biol Chem ; 287(16): 12679-90, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22294697

ABSTRACT

Abcb6 is a mammalian mitochondrial ATP-binding cassette (ABC) transporter that regulates de novo porphyrin synthesis. In previous studies, haploinsufficient (Abcb6(+/-)) embryonic stem cells showed impaired porphyrin synthesis. Unexpectedly, Abcb6(-/-) mice derived from these stem cells appeared phenotypically normal. We hypothesized that other ATP-dependent and/or -independent mechanisms conserve porphyrins. Here, we demonstrate that Abcb6(-/-) mice lack mitochondrial ATP-driven import of coproporphyrin III. Gene expression analysis revealed that loss of Abcb6 results in up-regulation of compensatory porphyrin and iron pathways, associated with elevated protoporphyrin IX (PPIX). Phenylhydrazine-induced stress caused higher mortality in Abcb6(-/-) mice, possibly because of sustained elevation of PPIX and an inability to convert PPIX to heme despite elevated ferrochelatase levels. Therefore, Abcb6 is the sole ATP-dependent porphyrin importer, and loss of Abcb6 produces up-regulation of heme and iron pathways necessary for normal development. However, under extreme demand for porphyrins (e.g. phenylhydrazine stress), these adaptations appear inadequate, which suggests that under these conditions Abcb6 is important for optimal survival.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Mitochondria/metabolism , Oxidants/toxicity , Phenylhydrazines/toxicity , Porphyrins/metabolism , ATP-Binding Cassette Transporters/genetics , Adenosine Triphosphate/metabolism , Animals , Coproporphyrins/metabolism , Erythropoiesis/physiology , Female , Gene Expression/physiology , Heme/metabolism , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Knockout , Oxidative Stress/drug effects , Oxidative Stress/physiology , Protoporphyrins/metabolism
6.
Cancer Res ; 71(5): 1781-91, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21245102

ABSTRACT

Purine nucleoside antimetabolites, such as clofarabine, are effective antileukemic agents. However, their effectiveness depends on an initial activation step in which they are monophosphorylated by deoxycytidine kinase (dCK). Some purine nucleoside antimetabolites and their monophosphate derivatives are exported by the ABC transporter ABCG2. Because clofarabine is a dCK substrate, and we show substantial variation in dCK and ABCG2 in myeloid leukemia, we hypothesized that the activity of dCK may modulate ABCG2-mediated resistance to clofarabine by regulating the formation of clofarabine monophosphate. We show that ABCG2 influence on clofarabine cytotoxicity was markedly influenced by dCK activity. When dCK expression was reduced by siRNA, clofarabine cytotoxicity was strongly reduced by enhanced ABCG2-mediated efflux. Conversely, dCK overexpression blunted ABCG2-mediated efflux of clofarabine by increasing the formation of clofarabine nucleotides. The use of an ABCG2 inhibitor confirmed that ABCG2 export of clofarabine is maximal when dCK levels are minimal. Analysis of intracellular clofarabine metabolites suggested that ABCG2 exported clofarabine more readily than clofarabine monophosphate. That ABCG2 primarily effluxes clofarabine, but not chlorfarabine-monophosphate, was confirmed by HPLC analysis of drug exported from ABCG2-overexpressing cells. Because the level and function of dCK and ABCG2 vary substantially among other types of cancer, these findings have important implications not only for clofarabine therapy but for purine nucleoside therapy in general. Therefore, we propose that addition of ABCG2 inhibitors would effectively increase the antitumor efficacy of purine nucleosides by blocking drug efflux that may be a significant mode of resistance when dCK levels are low.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Adenine Nucleotides/metabolism , Antineoplastic Agents/metabolism , Arabinonucleosides/metabolism , Deoxycytidine Kinase/metabolism , Drug Resistance, Neoplasm/physiology , Neoplasm Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Cell Line, Tumor , Clofarabine , Humans , Immunoblotting , Mutagenesis, Site-Directed , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction
7.
Cancer Res ; 67(14): 6965-72, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17638908

ABSTRACT

The use of probe substrates and combinations of ATP-binding cassette (ABC) transporter knockout (KO) animals may facilitate the identification of common substrates between apparently unrelated ABC transporters. An unexpectedly low concentration of the purine nucleotide analogue, 9-(2-(phosphonomethoxy)ethyl)-adenine (PMEA), and up-regulation of Abcg2 in some tissues of the Mrp4 KO mouse prompted us to evaluate the possibility that Abcg2 might transport purine-derived drugs. Abcg2 transported and conferred resistance to PMEA. Moreover, a specific Abcg2 inhibitor, fumitremorgin C, both increased PMEA accumulation and reversed Abcg2-mediated PMEA resistance. We developed Mrp4 and Abcg2 double KO mice and used both single KOs of Abcg2 and Mrp4 mice to assess the role of these transporters in vivo. Abcg2 contributed to PMEA accumulation in a variety of tissues, but in some tissues, this contribution was only revealed by the concurrent absence of Mrp4. Abcg2 also transported and conferred resistance to additional purine analogues, such as the antineoplastic, 2-chloro-2'-deoxyadenosine (cladribine) and puromycin, a protein synthesis inhibitor that is often used as a dominant selectable marker. Purine analogues interact with ABCG2 by a site distinct from the prazosin binding site as shown by their inability to displace the substrate analogue and photoaffinity tag [(125)I]iodoarylazidoprazosin. These studies show that Abcg2, like Mrp4, transports and confers resistance to purine nucleoside analogues and suggest that these two transporters work in parallel to affect drug cytotoxicity and tissue distribution. This new knowledge will facilitate an understanding of how Abcg2 and Mrp4, separately and in combination, protect against purine analogue host toxicity as well as resistance to chemotherapy.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Drug Resistance, Neoplasm , Multidrug Resistance-Associated Proteins/physiology , Purines/chemistry , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Animals , Antineoplastic Agents/pharmacology , Biological Transport , Cell Line, Tumor , Female , Humans , Mice , Mice, Knockout , Spleen/cytology , Substrate Specificity , Tissue Distribution
8.
Nature ; 443(7111): 586-9, 2006 Oct 05.
Article in English | MEDLINE | ID: mdl-17006453

ABSTRACT

The movement of anionic porphyrins (for example, haem) across intracellular membranes is crucial to many biological processes, but their mitochondrial translocation and coordination with haem biosynthesis is not understood. Transport of porphyrins into isolated mitochondria is energy-dependent, as expected for the movement of anions into a negatively charged environment. ATP-binding cassette transporters actively facilitate the transmembrane movement of substances. We found that the mitochondrial ATP-binding cassette transporter ABCB6 is upregulated (messenger RNA and protein in human and mouse cells) by elevation of cellular porphyrins and postulated that ABCB6 has a function in porphyrin transport. We also predicted that ABCB6 is functionally linked to haem biosynthesis, because its mRNA is found in both human bone marrow and CD71+ early erythroid cells (by database searching), and because our results show that ABCB6 is highly expressed in human fetal liver, and Abcb6 in mouse embryonic liver. Here we demonstrate that ABCB6 is uniquely located in the outer mitochondrial membrane and is required for mitochondrial porphyrin uptake. After ABCB6 is upregulated in response to increased intracellular porphyrin, mitochondrial porphyrin uptake activates de novo porphyrin biosynthesis. This process is blocked when the Abcb6 gene is silenced. Our results challenge previous assumptions about the intracellular movement of porphyrins and the factors controlling haem biosynthesis.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Porphyrins/metabolism , Animals , Biological Transport , Cell Differentiation , Fetus/metabolism , Gene Expression Regulation , Heme/metabolism , Humans , Liver/metabolism , Mice , Mitochondrial Membrane Transport Proteins/metabolism , Porphyrins/biosynthesis , Protein Binding , Protoporphyrins/metabolism
9.
Pharmacogenet Genomics ; 16(6): 415-27, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16708050

ABSTRACT

Two recent screens for copy-number variations in the entire human genome found 12.4 gene copy number variations per person, including 2.5% of individuals with gains between 7q21.1 and 7q22.1, the chromosomal location of CYP3A4. CYP3A4 is involved in the metabolism of approximately 50% of all drugs, including many cancer chemotherapeutic agents. CYP3A4 gene copy was determined in DNA from 143 individuals: normal human livers, primary and secondary liver tumors, human hepatic cell lines, and immortalized cell lines representing eight ethnically diverse populations. CYP3A4 gene copy was normal in all but one sample, a primary human hepatocellular carcinoma cell line (TONG/HCC). Southern blots of TONG/HCC DNA revealed an approximate 10-fold increase in CYP3A and a corresponding increase in CYP3A mRNA expression and catalytic activity. Fluorescent in situ hybridization of TONG/HCC revealed specific amplification of the CYP3A4 gene on chromosome 7q21 but no amplification of the MDR1 gene that localizes 11.9 Mb upstream of CYP3A4. High resolution analysis of DNA copy number by comparative genomic hybridization confirmed amplification at 7q21.3-7q22. The amplicon spanned 1.7 Mb and contained 30 known genes, including the entire CYP3A locus. To determine whether CYP3A4 expression affected chemotherapeutic toxicity, LLC-PK1 cells were transduced with adenoviruses expressing CYP3A4 and P450 reductase. CYP3A4 conferred resistance to taxol, vinblastine and topotecan. These studies demonstrate that CYP3A4 copy number differences do not contribute to the normal variation in CYP3A4 expression. Tumors with increased CYP3A copy number (via amplification or increased chromosome 7q) would be expected to show reduced cytotoxicity to some chemotherapeutic drugs and potentially an increase in the outgrowth of drug resistant tumors.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cytochrome P-450 Enzyme System/genetics , DNA/genetics , Gene Dosage , Liver Neoplasms/genetics , Blotting, Southern , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Chromosomes, Human, Pair 7 , Cytochrome P-450 CYP3A , Gene Amplification , Humans , In Situ Hybridization, Fluorescence , Liver Neoplasms/pathology , Nucleic Acid Hybridization , RNA, Messenger/metabolism
10.
Mol Cell Biol ; 24(17): 7612-21, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15314169

ABSTRACT

The role of the multidrug resistance protein MRP4/ABCC4 in vivo remains undefined. To explore this role, we generated Mrp4-deficient mice. Unexpectedly, these mice showed enhanced accumulation of the anticancer agent topotecan in brain tissue and cerebrospinal fluid (CSF). Further studies demonstrated that topotecan was an Mrp4 substrate and that cells overexpressing Mrp4 were resistant to its cytotoxic effects. We then used new antibodies to discover that Mrp4 is unique among the anionic ATP-dependent transporters in its dual localization at the basolateral membrane of the choroid plexus epithelium and in the apical membrane of the endothelial cells of the brain capillaries. Microdialysis sampling of ventricular CSF demonstrated that localization of Mrp4 at the choroid epithelium is integral to its function in limiting drug penetration into the CSF. The topotecan resistance of cells overexpressing Mrp4 and the polarized expression of Mrp4 in the choroid plexus and brain capillary endothelial cells indicate that Mrp4 has a dual role in protecting the brain from cytotoxins and suggest that the therapeutic efficacy of central nervous system-directed drugs that are Mrp4 substrates may be improved by developing Mrp4 inhibitors.


Subject(s)
Antineoplastic Agents/cerebrospinal fluid , Antineoplastic Agents/pharmacology , Brain/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Topotecan/cerebrospinal fluid , Topotecan/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Brain/blood supply , Brain/drug effects , Capillaries/cytology , Capillaries/metabolism , Cerebrovascular Circulation , Choroid Plexus/cytology , Choroid Plexus/metabolism , Drug Resistance, Multiple , Humans , Mice , Mice, Knockout , Microcirculation , Multidrug Resistance-Associated Proteins/genetics , Rats , Topotecan/pharmacokinetics
11.
J Biol Chem ; 279(21): 22250-7, 2004 May 21.
Article in English | MEDLINE | ID: mdl-15004017

ABSTRACT

The ABC transporter, Mrp4, transports the sulfated steroid DHEA-s, and sulfated bile acids interact with Mrp4 with high affinity. Hepatic Mrp4 levels are low, but increase under cholestatic conditions. We therefore inferred that up-regulation of Mrp4 during cholestasis is a compensatory mechanism to protect the liver from accumulation of hydrophobic bile acids. We determined that the nuclear receptor CAR is required to coordinately up-regulate hepatic expression of Mrp4 and an enzyme known to sulfate hydroxy-bile acids and steroids, Sult2a1. CAR activators increased Mrp4 and Sult2a1 expression in primary human hepatocytes and HepG2, a human liver cell line. Sult2a1 was down-regulated in Mrp4-null mice, further indicating an inter-relation between Mrp4 and Sult2a1 gene expression. Based on the hydrophilic nature of sulfated bile acids and the Mrp4 capability to transport sulfated steroids, our findings suggest that Mrp4 and Sult2a1 participate in an integrated pathway mediating elimination of sulfated steroid and bile acid metabolites from the liver.


Subject(s)
Multidrug Resistance-Associated Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Sulfotransferases/metabolism , Transcription Factors/metabolism , Animals , Bile Acids and Salts/metabolism , Blotting, Western , Cell Line , Cholestasis , Constitutive Androstane Receptor , Enhancer Elements, Genetic , Gene Expression Regulation , Genes, Reporter , Genotype , Green Fluorescent Proteins , Hepatocytes/metabolism , Humans , Hydroxy Acids/metabolism , Immunohistochemistry , Ligands , Liver/cytology , Liver/metabolism , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Models, Genetic , Multidrug Resistance-Associated Proteins/genetics , NIH 3T3 Cells , Oligonucleotide Array Sequence Analysis , Oligonucleotides/chemistry , Protein Binding , RNA/metabolism , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction , Steroids/metabolism , Transcription, Genetic , Transfection , Up-Regulation
12.
Hum Mol Genet ; 12(2): 99-109, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12499391

ABSTRACT

Drug transporters are an important part of the defense of cells against cytotoxic agents. One major group of transporters is known as multidrug resistance associated proteins (MRP; ABCC gene family). The MRPs belong to the ATP binding cassette transporter superfamily. One family member, ABCC4 (also known as MRP4) functions as a cellular efflux pump for anti-HIV drugs, such as 9-(2-phoshoenylmethoxyethyl) adenine and azido-thymidine-monophosphate, an antiviral nucleotide, ganciclovir-monophosphate, and anti-cancer agents such as thiopurines. We isolated a ABCC4 cDNA encoding a non-functional protein, owing to an insertion, and subsequently determined the ABCC4 gene structure. This analysis revealed that the insertion was attributed to two additional exons that would be predicted to produce premature termination codons (PTC) in ABCC4. The highly similar mouse Abcc4 gene also contained these exons, which were remarkable because their size and sequence identity were much higher than the overall similarity between these genes. Further, a comparison of human, monkey and rodent ABCC4 genes revealed that these same PTC-producing exons were also highly conserved in evolution. As all the ABCC4 mRNA containing these PTC exons might produce nonsense mRNA, we further tested the hypothesis that these mRNAs were targets of nonsense-mediated mRNA decay (NMD). Protein synthesis inhibition selectively stabilized PTC containing ABCC4 transcripts in human, monkey and rodent cell lines. Moreover, the amount of PTC-containing ABCC4 transcripts was critically dependent upon protein synthesis, as removal of the inhibitor dramatically decreased expression, which correlated with the resumption of protein synthesis. These are the first studies to indicate that the highly conserved PTC exons of the ABCC4 gene may dictate its expression.


Subject(s)
Alternative Splicing/genetics , Codon, Nonsense/genetics , Codon, Terminator/genetics , Gene Expression Regulation , Multidrug Resistance-Associated Proteins/genetics , Animals , Base Sequence , Cell Line , Conserved Sequence , Down-Regulation , Exons , Haplorhini , Humans , Mice , Molecular Sequence Data , Mutation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic
13.
J Biol Chem ; 277(41): 38998-9004, 2002 Oct 11.
Article in English | MEDLINE | ID: mdl-12105214

ABSTRACT

The multidrug resistance protein MRP4, a member of the ATP-binding cassette superfamily, confers resistance to purine-based antiretroviral agents. However, the antiviral agent ganciclovir (GCV) has not been shown to be a substrate of MRP4. GCV is important not only in antiviral therapy, but also in the selective killing of tumor cells modified to express herpes simplex virus thymidine kinase (HSV-TK). We therefore tested the effect of MRP4 on the cytotoxicity of GCV, on the ability of GCV to kill cells genetically modified to express HSV-TK, and on the bystander effect in which unmodified target cells are killed by GCV. Cells overexpressing MRP4 had markedly increased resistance to the cytotoxicity of GCV. Although, expression of recombinant HSV-TK increased the intracellular concentration of GCV nucleotide, cells were rescued by the cytoprotective effect of MRP4. In cells that overexpressed MRP4, intracellular accumulation of GCV metabolites was reduced, efflux of these metabolites was increased, and resistance to bystander killing was increased. Therefore, MRP4 can strongly reduce the susceptibility of HSV-TK-expressing cells to GCV, and its overexpression in adjacent cells protects them from bystander cell death. These findings indicate that a nucleotide transporter, such as MRP4, modulates the cellular response to GCV and thus may influence not only the efficacy of antiviral therapy, but also prodrug-based gene therapy, which is critically dependent upon bystander cell killing.


Subject(s)
Bystander Effect/drug effects , Drug Resistance, Multiple , Ganciclovir/pharmacology , Multidrug Resistance-Associated Proteins/metabolism , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line , Ganciclovir/metabolism , Humans , Molecular Sequence Data , Multidrug Resistance-Associated Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Simplexvirus/enzymology , Simplexvirus/genetics , T-Lymphocytes/metabolism , Thymidine Kinase/genetics , Thymidine Kinase/metabolism
14.
Biochim Biophys Acta ; 1574(2): 117-30, 2002 Mar 19.
Article in English | MEDLINE | ID: mdl-11955620

ABSTRACT

We previously reported on the isolation of a new rat ATP binding cassette (ABC) transporter, ABCB6. We now report the isolation of the full-length cDNA and genomic clones containing the human ABCB6 gene. ABCB6 is 100% identical to the cloned MTABC3 human ABC transporter and contains the typical ABC signature, Walker A and B motifs. We found that HuABCB6 is expressed at low levels in normal human liver. We found that ABCB6 was overexpressed in human hepatocellular carcinomas compared to paired surrounding non-malignant tissue. We found that there was no difference in ABCB6 gene copy between human liver cancer and its paired non-malignant tissue. Because HuABCB6 was overexpressed in human cancers compared to peri-tumoral tissue in the absence of gene amplification, transcriptional regulation may play an important role in its expression. Therefore, we isolated a 14 kb genomic DNA clone containing the HuABCB6 promoter and 5'-flanking region. The 5'-flanking region contains a CpG island, lacks an appropriately positioned TATA element and contains a number of putative transcription factor binding sites. Two transcription start sites were identified by S1 nuclease mapping at -274 and -296 bp from the start codon. Transient transfection of the HuABCB6 promoter constructs (HuABCB6/1.68, 1.39, 1.13, 0.90, 0.52) containing the luciferase reporter gene resulted in a 1100-2300-fold increase in luciferase activity compared to the empty vector control whereas HuABCB6/1.68 subcloned in the reverse orientation resulted in no activity. We observed a significant decrease in luciferase activity with the promoter constructs, HuABCB6/0.25, 0.15 and 0.06, which indicates that an orientation-dependent functional promoter is contained within our previously predicted promoter region of -315 bp to -565 bp as deletion of this 250 bp sequence resulted in a loss of promoter activity.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/chemistry , Amino Acid Sequence , Base Sequence , Binding Sites , Cloning, Molecular , DNA, Complementary/biosynthesis , DNA, Complementary/chemistry , Humans , Liver/metabolism , Molecular Sequence Data , Promoter Regions, Genetic , RNA, Messenger/metabolism , Single-Strand Specific DNA and RNA Endonucleases , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...