Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
J Neurol ; 253(2): 231-6, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16184340

ABSTRACT

Apoptotic deletion of autoreactive T-cells is defective in patients with multiple sclerosis (MS). Glatiramer acetate (GA) treatment seems to restore apoptosis of detrimental T-cells. We analyzed the mitochondria membrane pro- (Bax) and anti-apoptotic (Bcl- 2) and cytosolic pro-apoptotic (Cyt-c, APAF-1) proteins expression in peripheral lymphocytes from relapsing-remitting (RR) MS patients during GA treatment. Blood samples were collected from 8 healthy controls (HCs) and from 8 RR MS patients prior to and every three months during the 9 months of GA treatment. Peripheral blood mononuclear cells (PBMNCs) Bcl-2, Bax, Cyt-c and APAF-1 were quantified by western blot followed by densitometric scanning and Bax/Bcl-2, cytosolic Cyt-c/Bcl-2 and APAF-1/Bcl-2 ratios were calculated. T-cells were in vitro tested for oxygen consumption by a respirometric analysis. Bax/Bcl-2, cytosolic Cyt-c/Bcl-2 and APAF-1/Bcl-2 ratios in untreated MS patients were significantly (p < 0.05) lower than in HCs. Bax/Bcl-2 ratio increased (p = 0.03) and Cyt-c/Bcl-2 ratio showed a trend to increase during the 9 months of GA treatment in MS patients. A reduction of 58% and 59% in oxygen consumption by PBMNCs was evident after GA treatment in vitro or when GA treated patients' cells were compared with those from HCs, respectively. Our findings suggest that GA exerts a regulatory effect on peripheral T lymphocytes through pro-apoptosis mechanisms involving mitochondria and cytosolic proteins.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Immunosuppressive Agents/administration & dosage , Lymphocytes/drug effects , Multiple Sclerosis/pathology , Peptides/administration & dosage , Adult , Apoptotic Protease-Activating Factor 1 , Blotting, Western/methods , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Case-Control Studies , Cytochromes c/metabolism , Female , Glatiramer Acetate , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lymphocytes/pathology , Male , Middle Aged , Multiple Sclerosis/drug therapy , Oxygen Consumption/drug effects , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rotenone/pharmacology , Statistics, Nonparametric , Time Factors , Uncoupling Agents/pharmacology , bcl-2-Associated X Protein/metabolism
2.
J Biol Chem ; 276(52): 49053-60, 2001 Dec 28.
Article in English | MEDLINE | ID: mdl-11598111

ABSTRACT

MDR1 (multidrug resistance) P-glycoprotein (Pgp; ABCB1) decreases intracellular concentrations of structurally diverse drugs. Although Pgp is generally thought to be an efflux transporter, the mechanism of action remains elusive. To determine whether Pgp confers drug resistance through changes in transmembrane potential (E(m)) or ion conductance, we studied electrical currents and drug transport in Pgp-negative MCF-7 cells and MCF-7/MDR1 stable transfectants that were established and maintained without chemotherapeutic drugs. Although E(m) and total membrane conductance did not differ between MCF-7 and MCF-7/MDR1 cells, Pgp reduced unidirectional influx and steady-state cellular content of Tc-Sestamibi, a substrate for MDR1 Pgp, without affecting unidirectional efflux of substrate from cells. Depolarization of membrane potentials with various concentrations of extracellular K(+) in the presence of valinomycin did not inhibit the ability of Pgp to reduce intracellular concentration of Tc-Sestamibi, strongly suggesting that the drug transport activity of MDR1 Pgp is independent of changes in E(m) or total ion conductance. Tetraphenyl borate, a lipophilic anion, enhanced unidirectional influx of Tc-Sestamibi to a greater extent in MCF-7/MDR1 cells than in control cells, suggesting that Pgp may, directly or indirectly, increase the positive dipole potential within the plasma membrane bilayer. Overall, these data demonstrate that changes in E(m) or macroscopic conductance are not coupled with function of Pgp in multidrug resistance. The dominant effect of MDR1 Pgp in this system is reduction of drug influx, possibly through an increase in intramembranous dipole potential.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Biological Transport/physiology , Drug Resistance, Multiple/physiology , Membrane Potentials/physiology , Adenocarcinoma , Breast Neoplasms , Cell Membrane/metabolism , Female , Humans , Ionophores/pharmacology , Organotechnetium Compounds/metabolism , Patch-Clamp Techniques , Tumor Cells, Cultured , Valinomycin/pharmacology
3.
Cancer Res ; 61(17): 6540-7, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11522652

ABSTRACT

IRF9/p48/ISGF3gamma (IRF9) is an IFN regulatory factor that mediates signaling by type I IFNs (IFNalpha and IFNbeta). After single-step selection of breast adenocarcinoma cells in paclitaxel, differential display and single gene analysis demonstrated that transcriptional activation of IRF9 and other IFN-responsive genes, independent of IFN, corresponded with resistance to antimicrotubule agents. Transient overexpression of IRF9 reproduced the drug-resistance phenotype and induced expression of IFN-responsive genes. However, drug resistance was not induced by overexpression of Stat1 or Stat2, or treatment with IFNalpha per se. Using a donor-matched array of cDNA prepared from human tumor and normal tissue from a variety of organs, we observed overexpression of IRF9 in approximately one-half of breast and uterine tumors, which indicated that IRF9 may be important in signaling in these tumor types. These data identify a novel IFN-independent role for IRF9 in the development of resistance to antimicrotubule agents in breast tumor cells and may link downstream mediators of IFN signaling to drug resistance in human cancers.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , DNA-Binding Proteins/physiology , Paclitaxel/pharmacology , Transcription Factors/physiology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Cycle/drug effects , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/physiology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Interferon Type I/physiology , Interferon-Stimulated Gene Factor 3 , Interferon-Stimulated Gene Factor 3, gamma Subunit , Microtubules/drug effects , Signal Transduction/physiology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transfection , Tumor Cells, Cultured , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism , Vinblastine/pharmacology
4.
Bioconjug Chem ; 11(6): 762-71, 2000.
Article in English | MEDLINE | ID: mdl-11087323

ABSTRACT

Rapid and efficient delivery of radioactive metal complexes to the cell interior would enable novel applications in medical imaging and radiotherapy. Membrane permeant peptide conjugates incorporating HIV-1 Tat transactivation protein sequences (GRKKRRQRRR) and an appropriate peptide-based motif (epsilon-KGC) that provides an N(3)S donor core for chelating technetium and rhenium were synthesized. Oxotechnetium(V) and oxorhenium(V) Tat-peptide complexes were prepared by facile transchelation reactions with permetalates, tin(II) chloride and sodium glucoheptonate. RP-HPLC showed two major [(99m)Tc]Tat-peptide species (4) that differed in retention time by approximately 2 min corresponding to two [Re]Tat-peptide species (7) shown to have identical mass, consistent with formation of two isomers, likely the oxo-metal diastereomers. [(99m)Tc]Tat-peptides were stable to transchelation in vitro. In human Jurkat cells, [(99m)Tc]Tat-peptide 4 showed concentrative cell accumulation (30-fold greater than extracellular concentration) and rapid uptake kinetics (t(1/2) < 2 min) in a diastereomeric-comparable manner. Paradoxically, uptake was enhanced in 4 degrees C buffer compared to 37 degrees C, while depolarization of membrane potential as well as inhibition of microtubule function and vesicular trafficking showed no inhibitory effect. Cells preloaded with 4 showed rapid washout kinetics into peptide-free solution. Modification of [(99m)Tc]Tat-peptide by deletion of the N-terminus Gly with or without biotinylation minimally impacted net cell uptake. In addition, the C-terminus thiol of the prototypic Tat-peptide was labeled with fluorescein-5-maleimide to yield conjugate 8. Fluorescence microscopy directly localized conjugate 8 to the cytosol and nuclei (possibly nucleolus) of human Jurkat, KB 3-1 and KB 8-5 tumor cells. Preliminary imaging studies in mice following intravenous administration of prototypic [(99m)Tc]Tat-peptide 4 showed an initial whole body distribution and rapid clearance by both renal and hepatobiliary excretion. Analysis of murine blood in vivo and human serum ex vivo revealed >95% intact complex, while murine urine in vivo showed 65% parent complex. Thus, these novel Tat-peptide chelate conjugates, capable of forming stable [Tc/Re(V)]complexes, rapidly translocate across cell membranes into intracellular compartments and can be readily derivatized for further targeted applications in molecular imaging and radiotherapy.


Subject(s)
Chelating Agents/metabolism , Gene Products, tat/chemistry , Organotechnetium Compounds/metabolism , Peptide Fragments/metabolism , Radiopharmaceuticals/metabolism , Radiotherapy , Rhenium/metabolism , Amino Acid Sequence , Animals , Chelating Agents/chemistry , HIV-1/metabolism , Humans , Mice , Mice, Inbred BALB C , Organotechnetium Compounds/pharmacokinetics , Organotechnetium Compounds/therapeutic use , Peptide Fragments/chemistry , Peptide Fragments/pharmacokinetics , Radiopharmaceuticals/therapeutic use , Rhenium/pharmacokinetics , Rhenium/therapeutic use , Tissue Distribution , tat Gene Products, Human Immunodeficiency Virus
5.
Biochem Pharmacol ; 60(3): 413-26, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10856437

ABSTRACT

Multidrug resistance (MDR1) P-glycoprotein (Pgp), multidrug resistance-associated protein (MRP1), and breast cancer resistance protein (BCRP/MXR/ABCP) are members of the ATP-binding-cassette (ABC) superfamily of membrane transporters and are thought to function as energy-dependent efflux pumps of a variety of structurally diverse chemotherapeutic agents. We herein report the characterization of (99m)Tc-Tetrofosmin, a candidate radiopharmaceutical substrate of ABC transporters. (99m)Tc-Tetrofosmin showed high membrane potential-dependent accumulation in drug-sensitive KB 3-1 cells and low antagonist-reversible accumulation in MDR KB 8-5 and KB 8-5-11 cells in proportion to levels of MDR1 Pgp expression. In KB 8-5 cells, EC(50) values of the potent MDR antagonists N-(4-[2-(1,2,3, 4-tetrahydro-6,7-dimethoxy-2-isoquinolinyl)ethyl]-phenyl)-9, 10-dihydro-5-methoxy-9-oxo-4-acridine carboxamide (GF120918), (2R)-anti-5-¿3-[4-(10, 11-difluoromethanodibenzo-suber-5-yl)piperazin-1-yl]-2 -hydroxypropoxy ¿quinoline trihydrochloride (LY335979), and (3'-keto-Bmt')-[Val(2)]-cyclosporin A (PSC 833) were 40, 66, and 986 nM, respectively. Furthermore, only baculoviruses carrying human MDR1, but not MDR3, conferred both a decrease in accumulation of (99m)Tc-Tetrofosmin in host Spodoptera frugiperda (Sf9) cells and a GF120918-induced enhancement. Transport studies with a variety of stably transfected and drug-selected tumor cell lines were performed with (99m)Tc-Tetrofosmin and compared with (99m)Tc-Sestamibi, a previously validated MDR imaging agent. MDR1 Pgp readily transported each agent. To a lesser extent, MRP1 also transported each agent, likely as co-transport substrates with GSH; neither agent was a substrate for the BCRP/MXR/ABCP half-transporter. In mdr1a(-/-) and mdr1a/1b(-/-) mice, (99m)Tc-Tetrofosmin showed approximately 3. 5-fold greater brain uptake and retention compared with wild-type, with no net change in blood pharmacokinetics, consistent with transport in vivo by Pgp expressed at the capillary blood-brain barrier. Molecular imaging of the functional transport activity of ABC transporters in vivo with (99m)Tc-Tetrofosmin and related radiopharmaceuticals may enable non-invasive monitoring of chemotherapeutic and MDR gene therapy protocols.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/metabolism , Neoplasm Proteins , Organophosphorus Compounds/metabolism , Organotechnetium Compounds/metabolism , 3T3 Cells , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Animals , Baculoviridae/physiology , Biological Transport , Blood-Brain Barrier , Cross Reactions , Humans , KB Cells , Mice , Radiopharmaceuticals/metabolism , Reproducibility of Results , Subcellular Fractions , Tumor Cells, Cultured
6.
Biochemistry ; 39(26): 7651-61, 2000 Jul 04.
Article in English | MEDLINE | ID: mdl-10869171

ABSTRACT

Multidrug resistance P-glycoprotein (Pgp) has been reported to localize in low-density, cholesterol-enriched membranes. However, effects of low-density membrane domains on function of Pgp remain unexplored in whole cell systems. In cells that express modest levels of the protein endogenously or through drug selection, Pgp predominantly localized to low-density membranes following separation on a sucrose gradient. When highly overexpressed in NIH 3T3 cells, a prominent amount of Pgp also was detected in high-density membranes. Removing cholesterol from cells with beta-methylcyclodextrin (CD), a sterol acceptor molecule, shifted fractions that contained Pgp from low toward high density, and this effect was reversed to a similar extent by restoring sterols with either cholesterol or enantiomeric cholesterol. However, function of human MDR1 Pgp as probed with Tc-Sestamibi, a transport substrate for Pgp, was not dependent on localization of Pgp in cholesterol-enriched membranes. Specific inhibition of MDR1 Pgp with GF120918 or LY335979 also was independent of cholesterol. Cell-type-specific effects of cholesterol content on function of human Pgp were detected by use of daunomycin, another substrate for Pgp, although efficacy of inhibitors remained independent of cholesterol. Conversely, both function and inhibition of hamster Pgp as measured with Tc-Sestamibi and daunomycin were in part dependent on normal cell content of cholesterol. These data show that Pgp preferentially localizes to low-density, cholesterol-enriched membrane domains, but acute depletion of cholesterol impacts Pgp-mediated drug transport in a substrate- and cell-type-specific manner.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cholesterol, LDL/metabolism , Cholesterol/pharmacology , 3T3 Cells , ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Cholesterol/chemistry , Cholesterol, LDL/chemistry , Cholesterol, LDL/drug effects , Daunorubicin/metabolism , Humans , Mice , Microscopy, Fluorescence , Stereoisomerism , Technetium Tc 99m Sestamibi/metabolism
7.
Chem Biol ; 7(5): 335-43, 2000 May.
Article in English | MEDLINE | ID: mdl-10801474

ABSTRACT

BACKGROUND: Multidrug resistance (MDR) mediated by expression of MDR1 P-glycoprotein (Pgp) represents one of the best characterized barriers to chemotherapy in cancer patients. Positron emission tomography (PET) agents for analysis of Pgp-mediated drug transport activity in vivo would enable noninvasive assessment of chemotherapeutic regimens and MDR gene therapy. RESULTS: Candidate Schiff-base phenolic gallium(III) complexes were synthesized from their heptadentate precursors and gallium(III)acetylacetonate. Crystal structures demonstrated a hexacoordinated central gallium with overall trans-pseudo-octahedral geometry. Radiolabeled (67)Ga-complexes were obtained in high purity and screened in drug-sensitive (Pgp(-)) and MDR (Pgp(+)) tumor cells. Compared with control, lead compound 6. demonstrated antagonist-reversible 55-fold lower accumulation in Pgp-expressing MDR cells. Futhermore, compared with wild-type control, quantitative pharmacokinetic analysis showed markedly increased penetration and retention of 6. in brain and liver tissues of mdr1a/b((-/-)) gene disrupted mice, correctly mapping Pgp-mediated transport activity at the capillary blood-brain barrier and hepatocellular biliary cannalicular surface in vivo. CONCLUSIONS: These results indicate that gallium(III) complex 6. is recognized by MDR1 Pgp as an avid transport substrate, thereby providing a useful scaffold to generate (68)Ga radiopharmaceuticals for molecular imaging of Pgp transport activity in tumors and tissues in vivo using PET.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/pharmacokinetics , Gallium Radioisotopes/pharmacokinetics , Organometallic Compounds/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Biological Availability , Biological Transport , Humans , KB Cells , Mice , Mice, Knockout , Tomography, Emission-Computed , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...