Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Int J Mol Sci ; 25(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39062995

ABSTRACT

Breast cancer, a complex disease with a significant prevalence to form metastases, necessitates novel therapeutic strategies to improve treatment outcomes. Here, we present the results of a comparative molecular study of primary breast tumours, their metastases, and the corresponding primary cell lines using Desorption Electrospray Ionisation (DESI) and Laser-Assisted Rapid Evaporative Ionisation Mass Spectrometry (LA-REIMS) imaging. Our results show that ambient ionisation mass spectrometry technology is suitable for rapid characterisation of samples, providing a lipid- and metabolite-rich spectrum within seconds. Our study demonstrates that the lipidomic fingerprint of the primary tumour is not significantly distinguishable from that of its metastasis, in parallel with the similarity observed between their respective primary cell lines. While significant differences were observed between tumours and the corresponding cell lines, distinct lipidomic signatures and several phospholipids such as PA(36:2), PE(36:1), and PE(P-38:4)/PE(O-38:5) for LA-REIMS imaging and PE(P-38:4)/PE(O-38:5), PS(36:1), and PI(38:4) for DESI-MSI were identified in both tumours and cells. We show that the tumours' characteristics can be found in the corresponding primary cell lines, offering a promising avenue for assessing tumour responsiveness to therapeutic interventions. A comparative analysis by DESI-MSI and LA-REIMS imaging revealed complementary information, demonstrating the utility of LA-REIMS in the molecular imaging of cancer.


Subject(s)
Breast Neoplasms , Mammary Neoplasms, Animal , Cats , Animals , Female , Dogs , Cell Line, Tumor , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Cat Diseases/pathology , Spectrometry, Mass, Electrospray Ionization/methods , Neoplasm Metastasis , Dog Diseases/pathology , Dog Diseases/metabolism , Lipidomics/methods
2.
Int J Mol Sci ; 23(18)2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36142507

ABSTRACT

The multidrug transporter ABCB1 (MDR1, Pgp) plays an important role in the absorption, distribution, metabolism, and elimination of a wide range of pharmaceutical compounds. Functional investigation of the ABCB1 expression is also essential in many diseases, including drug-resistant cancer, inflammatory conditions, or Alzheimer disease. In this study, we examined the potential interaction of the ABCB1 multidrug transporter with a group of commercially available viability dyes that are generally considered not to penetrate into intact cells. Here, we demonstrate that the slow cellular accumulation of TO-PRO™-1 (TP1) or TO-PRO™-3 (TP3) was strongly inhibited by ABCB1-dependent dye extrusion. TP1/3 dye accumulation was not affected by the presence of ABCC1 or ABCG2, while this uptake was increased to the level in the ABCB1-negative cells by a specific P-glycoprotein inhibitor, Tariquidar. We suggest that TP compounds can be used as highly sensitive, selective, non-toxic, and stable dyes to examine the functional expression and properties of the ABCB1 multidrug transporter, especially in microplate-based high-throughput flow cytometry assays. In addition, we demonstrate the applicability of the TP dyes to efficiently select and separate even a very low number of Pgp-expressing intact cells.


Subject(s)
Fluorescent Dyes , Neoplasm Proteins , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Fluorescent Dyes/metabolism , Humans , Neoplasm Proteins/metabolism , Pharmaceutical Preparations
3.
Cell Mol Life Sci ; 76(20): 4131-4144, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31053883

ABSTRACT

ABCB6 belongs to the family of ATP-binding cassette (ABC) transporters, which transport various molecules across extra- and intra-cellular membranes, bearing significant impact on human disease and pharmacology. Although mutations in the ABCB6 gene have been linked to a variety of pathophysiological conditions ranging from transfusion incompatibility to pigmentation defects, its precise cellular localization and function is not understood. In particular, the intracellular localization of ABCB6 has been a matter of debate, with conflicting reports suggesting mitochondrial or endolysosomal expression. ABCB6 shows significant sequence identity to HMT-1 (heavy metal tolerance factor 1) proteins, whose evolutionarily conserved role is to confer tolerance to heavy metals through the intracellular sequestration of metal complexes. Here, we show that the cadmium-sensitive phenotype of Schizosaccharomyces pombe and Caenorhabditis elegans strains defective for HMT-1 is rescued by the human ABCB6 protein. Overexpression of ABCB6 conferred tolerance to cadmium and As(III) (As2O3), but not to As(V) (Na2HAsO4), Sb(V), Hg(II), or Zn(II). Inactivating mutations of ABCB6 abolished vacuolar sequestration of cadmium, effectively suppressing the cadmium tolerance phenotype. Modulation of ABCB6 expression levels in human glioblastoma cells resulted in a concomitant change in cadmium sensitivity. Our findings reveal ABCB6 as a functional homologue of the HMT-1 proteins, linking endolysosomal ABCB6 to the highly conserved mechanism of intracellular cadmium detoxification.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Cadmium/toxicity , Caenorhabditis elegans Proteins/genetics , Inactivation, Metabolic/genetics , Water Pollutants, Chemical/toxicity , ATP-Binding Cassette Transporters/deficiency , ATP-Binding Cassette Transporters/metabolism , Animals , Antimony/toxicity , Arsenates/toxicity , Arsenic Trioxide/toxicity , Cadmium/metabolism , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cell Line, Tumor , Conserved Sequence , Gene Expression , Genetic Complementation Test , HeLa Cells , Humans , Mercury/toxicity , Mutation , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Schizosaccharomyces/drug effects , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Vacuoles/drug effects , Vacuoles/metabolism , Water Pollutants, Chemical/metabolism , Zinc/toxicity
4.
Int J Mol Sci ; 21(4)2020 Feb 11.
Article in English | MEDLINE | ID: mdl-32053991

ABSTRACT

Breast cancer is the most commonly occurring cancer in women and the second most common cancer overall. By the age of 80, the estimated risk for breast cancer for women with germline BRCA1 or BRCA2 mutations is around 80%. Genetically engineered BRCA1-deficient mouse models offer a unique opportunity to study the pathogenesis and therapy of triple negative breast cancer. Here we present a newly established Brca1-/-, p53-/- mouse mammary tumor cell line, designated as CST. CST shows prominent features of BRCA1-mutated triple-negative breast cancers including increased motility, high proliferation rate, genome instability and sensitivity to platinum chemotherapy and PARP inhibitors (olaparib, veliparib, rucaparib and talazoparib). Genomic instability of CST cells was confirmed by whole genome sequencing, which also revealed the presence of COSMIC (Catalogue of Somatic Mutations in Cancer) mutation signatures 3 and 8 associated with homologous recombination (HR) deficiency. In vitro sensitivity of CST cells was tested against 11 chemotherapy agents. Tumors derived from orthotopically injected CST-mCherry cells in FVB-GFP mice showed sensitivity to cisplatin, providing a new model to study the cooperation of BRCA1-KO, mCherry-positive tumor cells and the GFP-expressing stromal compartment in therapy resistance and metastasis formation. In summary, we have established CST cells as a new model recapitulating major characteristics of BRCA1-negative breast cancers.


Subject(s)
BRCA1 Protein/genetics , Mammary Neoplasms, Animal/genetics , Triple Negative Breast Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Female , Gene Deletion , Genomic Instability , Mammary Neoplasms, Animal/pathology , Mice , Triple Negative Breast Neoplasms/pathology
5.
Arch Toxicol ; 93(4): 953-964, 2019 04.
Article in English | MEDLINE | ID: mdl-30863990

ABSTRACT

Membrane transporters play an important role in the absorption, distribution, metabolism and excretion of drugs. The cellular accumulation of many drugs is the result of the net function of efflux and influx transporters. Efflux transporters such as P-glycoprotein/ABCB1 have been shown to confer multidrug resistance in cancer. Although expression of uptake transporters has been confirmed in cancer cells, their role in chemotherapy response has not been systematically investigated. In the present study we have adapted a fluorescence-based cytotoxic assay to characterize the influence of key drug-transporters on the toxicity of approved anticancer drugs. Co-cultures of fluorescently labeled parental and transporter-expressing cells (expressing ABCB1, ABCG2 or OATP2B1) were screened against 101 FDA-approved anticancer drugs, using a novel, automated, triple fluorescence-based cytotoxicity assay. By measuring the survival of parental and transporter-expressing cells in co-cultures, we identify those FDA-approved anticancer drugs, whose toxicity is influenced by ABCB1, ABCG2 or OATP2B1. In addition to confirming known substrates of ABCB1 and ABCG2, the fluorescence-based cytotoxicity assays identified anticancer agents whose toxicity was increased in OATP2B1 expressing cells. Interaction of these compounds with OATP2B1 was verified in dedicated transport assays using cell-impermeant fluorescent substrates. Understanding drug-transporter interactions is needed to increase the efficacy of chemotherapeutic agents. Our results highlight the potential of the fluorescence-based HT screening system for identifying transporter substrates, opening the way for the design of therapeutic approaches based on the inhibition or even the exploitation of transporters in cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor/methods , Luminescent Proteins/genetics , Organic Anion Transporters/metabolism , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Coculture Techniques , Computer Simulation , Flow Cytometry , Fluorescent Dyes , Green Fluorescent Proteins/genetics , Humans , Organic Anion Transporters/genetics , Substrate Specificity , Transduction, Genetic , Red Fluorescent Protein
6.
Anal Chem ; 88(15): 7507-14, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27377867

ABSTRACT

Rapid evaporative ionization mass spectrometry (REIMS) was used for the rapid mass spectrometric profiling of cancer cell lines. Spectral reproducibility was assessed for three different cell lines, and the extent of interclass differences and intraclass variance was found to allow the identification of these cell lines based on the REIMS data. Subsequently, the NCI60 cell line panel was subjected to REIMS analysis, and the resulting data set was investigated for its distinction of individual cell lines and different tissue types of origin. Information content of REIMS spectral profiles of cell lines were found to be similar to those obtained from mammalian tissues although pronounced differences in relative lipid intensity were observed. Ultimately, REIMS was shown to detect changes in lipid content of cell lines due to mycoplasma infection. The data show that REIMS is an attractive means to study cell lines involving minimal sample preparation and analysis times in the range of seconds.


Subject(s)
Lipids/analysis , Cell Line, Tumor/microbiology , Humans , Mass Spectrometry/methods , Mycoplasma , Neoplasms/metabolism , Principal Component Analysis , Reproducibility of Results
7.
Biochem J ; 467(1): 127-39, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25627919

ABSTRACT

ATP-binding cassette, subfamily B (ABCB) 6 is a homodimeric ATP-binding cassette (ABC) transporter present in the plasma membrane and in the intracellular organelles. The intracellular localization of ABCB6 has been a matter of debate, as it has been suggested to reside in the mitochondria and the endo-lysosomal system. Using a variety of imaging modalities, including confocal microscopy and EM, we confirm the endo-lysosomal localization of ABCB6 and show that the protein is internalized from the plasma membrane through endocytosis, to be distributed to multivesicular bodies and lysosomes. In addition to the canonical nucleotide-binding domain (NBD) and transmembrane domain (TMD), ABCB6 contains a unique N-terminal TMD (TMD0), which does not show sequence homology to known proteins. We investigated the functional role of these domains through the molecular dissection of ABCB6. We find that the folding, dimerization, membrane insertion and ATP binding/hydrolysis of the core-ABCB6 complex devoid of TMD0 are preserved. However, in contrast with the full-length transporter, the core-ABCB6 construct is retained at the plasma membrane and does not appear in Rab5-positive endosomes. TMD0 is directly targeted to the lysosomes, without passage to the plasma membrane. Collectively, our results reveal that TMD0 represents an independently folding unit, which is dispensable for catalysis, but has a crucial role in the lysosomal targeting of ABCB6.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Endocytosis , Lysosomes/metabolism , Multivesicular Bodies/metabolism , Protein Sorting Signals , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , Adenosine Triphosphate/metabolism , Amino Acid Substitution , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Dimerization , HeLa Cells , Humans , Hydrolysis , K562 Cells , Lysosomes/ultrastructure , Microscopy, Electron, Transmission , Multivesicular Bodies/ultrastructure , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Folding , Protein Structure, Tertiary , Protein Transport , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
8.
J Med Chem ; 67(11): 9069-9090, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38771959

ABSTRACT

The development of copper(II) thiosemicarbazone complexes as potential anticancer agents, possessing dual functionality as inhibitors of R2 ribonucleotide reductase (RNR) and tubulin polymerization by binding at the colchicine site, presents a promising avenue for enhancing therapeutic effectiveness. Herein, we describe the syntheses and physicochemical characterization of four isomeric proligands H2L3-H2L6, with the methylmorpholine substituent at pertinent positions of the pyridine ring, along with their corresponding Cu(II) complexes 3-6. Evidently, the position of the morpholine moiety and the copper(II) complex formation have marked effects on the in vitro antiproliferative activity in human uterine sarcoma MES-SA cells and the multidrug-resistant derivative MES-SA/Dx5 cells. Activity correlated strongly with quenching of the tyrosyl radical (Y•) of mouse R2 RNR protein, inhibition of RNR activity in the cancer cells, and inhibition of tubulin polymerization. Insights into the mechanism of antiproliferative activity, supported by experimental results and molecular modeling calculations, are presented.


Subject(s)
Antineoplastic Agents , Copper , Morpholines , Ribonucleotide Reductases , Thiosemicarbazones , Tubulin , Thiosemicarbazones/chemistry , Thiosemicarbazones/pharmacology , Thiosemicarbazones/chemical synthesis , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Ribonucleotide Reductases/antagonists & inhibitors , Ribonucleotide Reductases/metabolism , Tubulin/metabolism , Animals , Morpholines/pharmacology , Morpholines/chemistry , Morpholines/chemical synthesis , Copper/chemistry , Mice , Cell Line, Tumor , Cell Proliferation/drug effects , Structure-Activity Relationship , Polymerization/drug effects , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Pyridines/pharmacology , Pyridines/chemistry , Pyridines/chemical synthesis , Tubulin Modulators/pharmacology , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry , Drug Screening Assays, Antitumor , Models, Molecular
9.
Front Pharmacol ; 13: 958023, 2022.
Article in English | MEDLINE | ID: mdl-36120371

ABSTRACT

Organic anion transporting polypeptide 3A1 (OATP3A1, encoded by the SLCO3A1 gene) is a prostaglandin, oligopeptide, and steroid/thyroid hormone transporter with wide tissue distribution, expressed, e.g., in the human brain and testis. Although the physiological importance of OATP3A1 has not yet been clarified, based on its expression pattern, substrate recognition, and evolutionary conservation, OATP3A1 is a potential pharmacological target. Previously, two isoforms of OATP3A1, termed as V1 and V2, have been characterized. Here, we describe the cloning and functional characterization of a third isoform, OATP3A1_V3. The mRNA of isoform V3 is formed by alternative splicing and results in an OATP3A1 protein with an altered C-terminus compared to isoforms V1 and V2. Based on quantitative PCR, we demonstrate the widespread expression of SLCO3A1_V3 mRNA in human organs, with the highest expression in the brain and testis. By generation of an isoform V3-specific antibody and immunostaining, we show that the encoded protein is expressed in the human choroid plexus, neurons, and both germ and Sertoli cells of the testis. Moreover, we demonstrate that in contrast to isoform V1, OATP3A1_V3 localizes to the apical membrane of polarized MDCKII cells. Using HEK-293 cells engineered to overexpress OATP3A1_V3, we verify the protein's functionality and identify dehydroepiandrosterone sulfate as a novel OATP3A1 substrate. Based on their distinct expression patterns but overlapping functions, OATP3A1 isoforms may contribute to transcellular (neuro)steroid transport in the central nervous system.

10.
Biology (Basel) ; 11(11)2022 Nov 16.
Article in English | MEDLINE | ID: mdl-36421383

ABSTRACT

The human GLUT1 (SLC2A1) membrane protein is the key glucose transporter in numerous cell types, including red cells, kidney, and blood-brain barrier cells. The expression level of this protein has a role in several diseases, including cancer and Alzheimer's disease. In this work, to investigate a potential genetic modulation of the GLUT1 expression level, the protein level was measured in red cell membranes by flow cytometry, and the genetic background was analyzed by qPCR and luciferase assays. We found significant associations between red cell GLUT1 levels and four single nucleotide polymorphisms (SNP) in the coding SLC2A1 gene, that in individuals with the minor alleles of rs841848, rs1385129, and rs11537641 had increased, while those having the variant rs841847 had decreased erythrocyte GLUT1 levels. In the luciferase reporter studies performed in HEK-293T and HepG2 cells, a similar SNP-dependent modulation was observed, and lower glucose, serum, and hypoxic condition had variable, cell- and SNP-specific effects on luciferase expression. These results should contribute to a more detailed understanding of the genetic background of membrane GLUT1 expression and its potential role in associated diseases.

11.
Cancers (Basel) ; 13(1)2021 Jan 05.
Article in English | MEDLINE | ID: mdl-33466433

ABSTRACT

Resistance to chemotherapeutic agents is a major obstacle in cancer treatment. A recently proposed strategy is to target the collateral sensitivity of multidrug resistant (MDR) cancer. Paradoxically, the toxicity of certain metal chelating agents is increased, rather than decreased, by the function of P-glycoprotein (Pgp), which is known to confer resistance by effluxing chemotherapeutic compounds from cancer cells. We have recently characterized and compared the solution's chemical properties including ligand protonation and the metal binding properties of a set of structurally related 8-hydroxyquinoline derived Mannich bases. Here we characterize the impact of the solution stability and redox activity of their iron(III) and copper(II) complexes on MDR-selective toxicity. Our results show that the MDR-selective anticancer activity of the studied 8-hydroxyquinoline derived Mannich bases is associated with the iron deprivation of MDR cells and the preferential formation of redox-active copper(II) complexes, which undergo intracellular redox-cycling to induce oxidative stress.

12.
Sci Rep ; 10(1): 19739, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33184408

ABSTRACT

The phospholipid biosynthesis of the malaria parasite, Plasmodium falciparum is a key process for its survival and its inhibition is a validated antimalarial therapeutic approach. The second and rate-limiting step of the de novo phosphatidylcholine biosynthesis is catalysed by CTP: phosphocholine cytidylyltransferase (PfCCT), which has a key regulatory function within the pathway. Here, we investigate the functional impact of the key structural differences and their respective role in the structurally unique pseudo-heterodimer PfCCT protein in a heterologous cellular context using the thermosensitive CCT-mutant CHO-MT58 cell line. We found that a Plasmodium-specific lysine-rich insertion within the catalytic domain of PfCCT acts as a nuclear localization signal and its deletion decreases the nuclear propensity of the protein in the model cell line. We further showed that the putative membrane-binding domain also affected the nuclear localization of the protein. Moreover, activation of phosphatidylcholine biosynthesis by phospholipase C treatment induces the partial nuclear-to-cytoplasmic translocation of PfCCT. We additionally investigated the cellular function of several PfCCT truncated constructs in a CHO-MT58 based rescue assay. In absence of the endogenous CCT activity we observed that truncated constructs lacking the lysine-rich insertion, or the membrane-binding domain provided similar cell survival ratio as the full length PfCCT protein.


Subject(s)
Cell Nucleus/metabolism , Choline-Phosphate Cytidylyltransferase/chemistry , Choline-Phosphate Cytidylyltransferase/metabolism , Nuclear Localization Signals , Plasmodium falciparum/enzymology , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Amino Acid Sequence , Animals , CHO Cells , Catalytic Domain , Cell Nucleus/genetics , Choline-Phosphate Cytidylyltransferase/genetics , Cricetinae , Cricetulus , Cytidine Triphosphate/metabolism , Phosphorylcholine/metabolism , Protein Binding , Sequence Homology, Amino Acid
13.
FEBS J ; 287(12): 2468-2485, 2020 06.
Article in English | MEDLINE | ID: mdl-31770475

ABSTRACT

Organic anion-transporting polypeptide 1A2 (OATP1A2), expressed in the human blood-brain barrier, promotes drug uptake from the blood and hence can be exploited for central nervous system-targeted drug delivery. The thyroid transporter OATP1C1, expressed in the choroid plexus and in astrocytes, is also a potential pharmacological target. Based on their established pharmacological relevance, screening the drug interaction profile of OATP1A2 and OATP1C1 is highly desirable. However, drug interaction screens require suitable model systems and functional assays. In the current study, uptake of a set of cell-impermeable fluorescent dyes was screened in HEK-293 and A431 cell lines overexpressing OATP1A2 and OATP1C1. Based on the uptake of fluorescent dye substrates, a functional assay was developed, which was used to characterize OATP inhibitors/substrates. We identify Live/Dead Green (LDG), Live-or-Dye 488, and sulforhodamines 101, G, and B as novel fluorescent substrates of OATP1A2 and OATP1C1. We show that LDG uptake is proportional to OATP1A2/1C1 expression, allowing the isolation of cells expressing high transporter levels. Additionally, dye uptake can be used to characterize the drug interaction pattern of OATP1A2 and OATP1C1. We demonstrate that third-generation P-glycoprotein inhibitors elacridar, tariquidar, and zosuquidar inhibit OATP1A2 function. Increased toxicity of elacridar in OATP1A2-expressing cells suggests that OATP1A2 may modulate the distribution of this compound. The fluorescence-based assays developed in the current study are a good alternative of radioligand-based tests and pave the way toward high-throughput screens for OATP1A2/1C1 drug interaction studies.


Subject(s)
Acridines/pharmacology , Dibenzocycloheptenes/pharmacology , Fluorescent Dyes/chemistry , Organic Anion Transporters/antagonists & inhibitors , Quinolines/pharmacology , Tetrahydroisoquinolines/pharmacology , Acridines/chemistry , Cell Line , Dibenzocycloheptenes/chemistry , Humans , Organic Anion Transporters/analysis , Organic Anion Transporters/metabolism , Quinolines/chemistry , Tetrahydroisoquinolines/chemistry
14.
Sci Rep ; 10(1): 8880, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32483232

ABSTRACT

Transmembrane proteins (TMP) play a crucial role in several physiological processes. Despite their importance and diversity, only a few TMP structures have been determined by high-resolution protein structure characterization methods so far. Due to the low number of determined TMP structures, the parallel development of various bioinformatics and experimental methods was necessary for their topological characterization. The combination of these methods is a powerful approach in the determination of TMP topology as in the Constrained Consensus TOPology prediction. To support the prediction, we previously developed a high-throughput topology characterization method based on primary amino group-labelling that is still limited in identifying all TMPs and their extracellular segments on the surface of a particular cell type. In order to generate more topology information, a new step, a partial proteolysis of the cell surface has been introduced to our method. This step results in new primary amino groups in the proteins that can be biotinylated with a membrane-impermeable agent while the cells still remain intact. Pre-digestion also promotes the emergence of modified peptides that are more suitable for MS/MS analysis. The modified sites can be utilized as extracellular constraints in topology predictions and may contribute to the refined topology of these proteins.


Subject(s)
Membrane Proteins/chemistry , Membrane Proteins/metabolism , Algorithms , Biotinylation , Databases, Protein , HL-60 Cells , Humans , Microscopy, Confocal , Protein Domains , Proteolysis , Staining and Labeling , Tandem Mass Spectrometry
15.
Cancer Res ; 80(4): 663-674, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31888888

ABSTRACT

Clinical evidence shows that following initial response to treatment, drug-resistant cancer cells frequently evolve and, eventually, most tumors become resistant to all available therapies. We compiled a focused library consisting of >500 commercially available or newly synthetized 8-hydroxyquinoline (8OHQ) derivatives whose toxicity is paradoxically increased rather than decreased by the activity of P-glycoprotein (Pgp), a transporter conferring multidrug resistance (MDR). Here, we deciphered the mechanism of action of NSC297366 that shows exceptionally strong Pgp-potentiated toxicity. Treatment of cells with NSC297366 resulted in changes associated with the activity of potent anticancer iron chelators. Strikingly, iron depletion was more pronounced in MDR cells due to the Pgp-mediated efflux of NSC297366-iron complexes. Our results indicate that iron homeostasis can be targeted by MDR-selective compounds for the selective elimination of multidrug resistant cancer cells, setting the stage for a therapeutic approach to fight transporter-mediated drug resistance. SIGNIFICANCE: Modulation of the MDR phenotype has the potential to increase the efficacy of anticancer therapies. These findings show that the MDR transporter is a "double-edged sword" that can be turned against resistant cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Iron Chelating Agents/pharmacology , Iron/metabolism , Neoplasms/drug therapy , Oxyquinoline/pharmacology , ATP Binding Cassette Transporter, Subfamily B/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Inhibitory Concentration 50 , Iron Chelating Agents/therapeutic use , Neoplasms/pathology , Oxyquinoline/analogs & derivatives , Oxyquinoline/therapeutic use
16.
Rapid Commun Mass Spectrom ; 23(21): 3372-6, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19780062

ABSTRACT

ATP-Binding Cassette (ABC) transporters are highly expressed in pharmacological barriers limiting the access of drugs to their targets. Since characterization of a compound as a transporter substrate or inhibitor bears significant consequences in drug development, there is a great need for reliable tools that enable the rapid analysis of the transport susceptibility of drugs. Here we describe a simple but very efficient high-performance liquid chromatography/mass spectrometry (HPLC/MS) assay for measuring the ABC transporter-dependent vesicular transport of compounds. In addition, we provide evidence that the requirement for sample preparation can be minimized using desorption electrospray ionization (DESI)-MS, paving the way for a direct, high-throughput investigation of drug-transporter interactions.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Chromatography, High Pressure Liquid/methods , Spectrometry, Mass, Electrospray Ionization/methods , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Humans , Methotrexate/pharmacokinetics , Neoplasm Proteins/metabolism , Pharmacokinetics , Transport Vesicles/metabolism
17.
Sci Rep ; 9(1): 15729, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31673029

ABSTRACT

The research on transmembrane proteins (TMPs) is quite widespread due to their biological importance. Unfortunately, only a little amount of structural data is available of TMPs. Since technical difficulties arise during their high-resolution structure determination, bioinformatics and other experimental approaches are widely used to characterize their low-resolution structure, namely topology. Experimental and computational methods alone are still limited to determine TMP topology, but their combination becomes significant for the production of reliable structural data. By applying amino acid specific membrane-impermeable labelling agents, it is possible to identify the accessible surface of TMPs. Depending on the residue-specific modifications, new extracellular topology data is gathered, allowing the identification of more extracellular segments for TMPs. A new method has been developed for the experimental analysis of TMPs: covalent modification of the carboxyl groups on the accessible cell surface, followed by the isolation and digestion of these proteins. The labelled peptide fragments and their exact modification sites are identified by nanoLC-MS/MS. The determined peptides are mapped to the primary sequences of TMPs and the labelled sites are utilised as extracellular constraints in topology predictions that contribute to the refined low-resolution structure data of these proteins.


Subject(s)
Carboxylic Acids/chemistry , Membrane Proteins/chemistry , Biotinylation , Cell Line, Tumor , Cell Membrane/metabolism , Chromatography, High Pressure Liquid , Cystamine/chemistry , Fluorescent Dyes/chemistry , Humans , Membrane Proteins/metabolism , Microscopy, Confocal , Nanotechnology , Peptide Fragments/analysis , Peptides/chemistry , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/metabolism , Tandem Mass Spectrometry
18.
Sci Rep ; 8(1): 2630, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29422623

ABSTRACT

Organic Anion-Transporting Polypeptides are multispecific membrane proteins that regulate the passage of crucial endobiotics and drugs across pharmacological barriers. OATP1B1 and OATP1B3 have been described to play a major role in the hepatic uptake of statins, antivirals and various chemotherapeutics; whereas the pharmacological role of the ubiquitously expressed OATP2B1 is less well characterized. According to current industry standards, in vitro testing for susceptibility to OATP1B1 and 1B3 mediated transport is recommended for drug candidates that are eliminated in part via the liver. Here we show that human OATP1B1, 1B3 and 2B1 transport a series of commercially available viability dyes that are generally believed to be impermeable to intact cells. We demonstrate that the intracellular accumulation of Zombie Violet, Live/Dead Green, Cascade Blue and Alexa Fluor 405 is specifically increased by OATPs. Inhibition of Cascade Blue or Alexa Fluor 405 uptake by known OATP substrates/inhibitors yielded IC50 values in agreement with gold-standard radioligand assays. The fluorescence-based assays described in this study provide a new tool for testing OATP1B/2B1 drug interactions.


Subject(s)
Liver-Specific Organic Anion Transporter 1/metabolism , Organic Anion Transporters/metabolism , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Animals , Biological Transport , Coloring Agents/metabolism , Dogs , Drug Interactions , HEK293 Cells , Humans , Liver/metabolism , Liver-Specific Organic Anion Transporter 1/antagonists & inhibitors , Madin Darby Canine Kidney Cells , Organic Anion Transporters/antagonists & inhibitors , Sf9 Cells , Solute Carrier Organic Anion Transporter Family Member 1B3/antagonists & inhibitors
19.
J Mol Biol ; 430(20): 3802-3818, 2018 10 12.
Article in English | MEDLINE | ID: mdl-29940187

ABSTRACT

Genetically inheritable pigmentation defects provide a unique opportunity to reveal the function of proteins contributing to melanogenesis. Dyschromatosis universalis hereditaria (DUH) is a rare pigmentary genodermatosis associated with mutations in the ABCB6 gene. Here we use optical and electron microscopy imaging combined with biochemical tools to investigate the localization and function of ABCB6 in pigment cells. We show that ABCB6 localizes to the membrane of early melanosomes and lysosomes of the human melanocytic cell line MNT-1. Depletion of ABCB6 by siRNA impaired PMEL amyloidogenesis in early melanosomes and induced aberrant accumulation of multilamellar aggregates in pigmented melanosomes. PMEL fibril formation and normal maturation of pigmented melanosomes could be restored by the overexpression of wild-type ABCB6 but not by variants containing an inactivating catalytic mutation (K629M) or the G579E DUH mutation. In line with the impairment of PMEL matrix formation in the absence of ABCB6, morphological analysis of the retinal pigment epithelium of ABCB6 knockout mice revealed a significant decrease of melanosome numbers. Our study extends the localization of ABCB6 to melanosomes, suggesting a potential link between the function of ABCB6 and the etiology of DUH to amyloid formation in pigment cells.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/metabolism , Amyloid/chemistry , Amyloid/metabolism , Melanosomes/metabolism , gp100 Melanoma Antigen/chemistry , gp100 Melanoma Antigen/metabolism , ATP-Binding Cassette Transporters/genetics , Animals , Cell Line , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Melanocytes/metabolism , Mice , Mice, Knockout , Protein Transport , RNA Interference , RNA, Small Interfering
20.
Sci Rep ; 8(1): 8932, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29895950

ABSTRACT

The plasmodial CTP:phosphocholine cytidylyltransferase (PfCCT) is a promising antimalarial target, which can be inhibited to exploit the need for increased lipid biosynthesis during the erythrocytic life stage of Plasmodium falciparum. Notable structural and regulatory differences of plasmodial and mammalian CCTs offer the possibility to develop species-specific inhibitors. The aim of this study was to use CHO-MT58 cells expressing a temperature-sensitive mutant CCT for the functional characterization of PfCCT. We show that heterologous expression of wild type PfCCT restores the viability of CHO-MT58 cells at non-permissive (40 °C) temperatures, whereas catalytically perturbed or structurally destabilized PfCCT variants fail to provide rescue. Detailed in vitro characterization indicates that the H630N mutation diminishes the catalytic rate constant of PfCCT. The flow cytometry-based rescue assay provides a quantitative readout of the PfCCT function opening the possibility for the functional analysis of PfCCT and the high throughput screening of antimalarial compounds targeting plasmodial CCT.


Subject(s)
Choline-Phosphate Cytidylyltransferase/metabolism , Phosphatidylcholines/biosynthesis , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Animals , Antimalarials/pharmacology , Biocatalysis/drug effects , Biosynthetic Pathways/drug effects , CHO Cells , Cell Survival/drug effects , Cell Survival/genetics , Choline-Phosphate Cytidylyltransferase/antagonists & inhibitors , Choline-Phosphate Cytidylyltransferase/genetics , Cricetinae , Cricetulus , Gene Expression Regulation, Enzymologic/drug effects , Mutation , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL