Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biomed Pharmacother ; 121: 109648, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31810115

ABSTRACT

Malignant cells frequently demonstrate an oncogenic-driven reliance on glycolytic metabolism to support their highly proliferative nature. Overexpression of pyruvate dehydrogenase kinase (PDK) may promote this unique metabolic signature of tumor cells by inhibiting mitochondrial function. PDKs function to phosphorylate and inhibit pyruvate dehydrogenase (PDH) activity. Silencing of PDK expression has previously been shown to restore mitochondrial function and reduce tumor cell proliferation. High dose Vitamin B1, or thiamine, possesses antitumor properties related to its capacity to reduce PDH phosphorylation and promote its enzymatic activity, presumably through PDK inhibition. Though a promising nutraceutical approach for cancer therapy, thiamine's low bioavailability may limit clinical effectiveness. Here, we have demonstrated exploiting the commercially available lipophilic thiamine analogs sulbutiamine and benfotiamine increases thiamine's anti-cancer effect in vitro. Determined by crystal violet proliferation assays, both sulbutiamine and benfotiamine reduced thiamine's millimolar IC50 value to micromolar equivalents. HPLC analysis revealed that sulbutiamine and benfotiamine significantly increased intracellular thiamine and TPP concentrations in vitro, corresponding with reduced levels of PDH phosphorylation. Through an ex vitro kinase screen, thiamine's activated cofactor form thiamine pyrophosphate (TPP) was found to inhibit the function of multiple PDK isoforms. Attempts to maximize intracellular TPP by exploiting thiamine homeostasis gene expression resulted in enhanced apoptosis in tumor cells. Based on our in vitro evaluations, we conclude that TPP serves as the active species mediating thiamine's inhibitory effect on tumor cell proliferation. Pharmacologic administration of benfotiamine, but not sulbutiamine, reduced tumor growth in a subcutaneous xenograft mouse model. It remains unclear if benfotiamine's effects in vivo are associated with PDK inhibition or through an alternative mechanism of action. Future work will aim to define the action of lipophilic thiamine mimetics in vivo in order to translate their clinical usefulness as anticancer strategies.


Subject(s)
Antineoplastic Agents/pharmacology , Dietary Supplements , Thiamine/analogs & derivatives , Thiamine/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Confidence Intervals , Female , Humans , Inhibitory Concentration 50 , Intracellular Space/metabolism , Mice, Nude , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Thiamine/chemistry , Thiamine Pyrophosphate/metabolism
2.
Article in English | MEDLINE | ID: mdl-31238261

ABSTRACT

Understanding the mechanisms underlying cancer cell survival is critical toward advancing drug discovery efforts in this field. Supplemental vitamins have been proposed to play a role in cancer cell metabolism because the increased supply of nutrients is thought to provide cofactors supporting the higher metabolic rate of cancer cells. Particularly, the role of thiamine (vitamin B1) in many biochemical pathways that supports cancer cell metabolism has been investigated. Consequently, the analysis of thiamine and its derivatives in a manner that reflects its dynamic response to genetic modification and pathophysiological stimuli is essential. In this work, we developed a mass spectrometry based-analytical method to track metabolites derived from stable isotope tracers for a better understanding of the metabolic fate of thiamine in cancer cells. This method used ion-pair reversed phase liquid chromatography to simultaneously quantify underivatized thiamine, thiamine monophosphate (TMP) and thiamine pyrophosphate (TPP) in cells. Hexylamine was used as an ion-pairing agent. The method was successfully validated for accuracy, precision and selectivity in accordance with U.S. FDA guidance. Furthermore, the method was then applied for the determination of thiamine and its derivatives with stable isotope labeling to explore the metabolic fate of intracellular thiamine in cancer cells. The finding shows that thiamine is rapidly converted to TPP however, the TPP does not return to thiamine. It appears that TPP may be utilized for other purposes rather than simply being an enzyme cofactor, suggesting unexplored roles for thiamine in cancer.


Subject(s)
Chromatography, High Pressure Liquid/methods , Tandem Mass Spectrometry/methods , Thiamine/analysis , Cell Line, Tumor , Humans , Thiamine/metabolism
3.
Oncotarget ; 9(83): 35422-35438, 2018 Oct 23.
Article in English | MEDLINE | ID: mdl-30459934

ABSTRACT

Supplemental levels of vitamin B1 (thiamine) have been implicated in tumor progression. Tumor cells adaptively up-regulate thiamine transport during hypoxic stress. Upon uptake, thiamine pyrophosphokinase-1 (TPK1) facilitates the rapid phosphorylation of thiamine into thiamine pyrophosphate (TPP). However, the regulation of TPK1 during hypoxic stress is undefined. Understanding how thiamine homeostasis changes during hypoxia will provide critical insight into the malignant advantage supplemental thiamine may provide cancer cells. Using Western blot analysis and RT-PCR, we have demonstrated the post-transcriptional up-regulation of TPK1 in cancer cells following hypoxic exposure. TPK1 expression was also adaptively up-regulated following alterations of redox status by chemotherapeutic and antioxidant treatments. Although TPK1 was functionally up-regulated by hypoxia, HPLC analysis revealed a reduction in intracellular TPP levels. This loss was reversed by treatment with cell-permeable antioxidants and corresponded with reduced ROS production and enhanced cellular proliferation during supplemental thiamine conditions. siRNA-mediated knockdown of TPK1 directly enhanced basal ROS levels and reduced tumor cell proliferation. These findings suggest that the adaptive regulation of TPK1 may be an essential component in the cellular response to oxidative stress, and that during supplemental thiamine conditions its expression may be exploited by tumor cells for a redox advantage contributing to tumor progression.

4.
Biomed Chromatogr ; 30(1): 35-41, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26120008

ABSTRACT

A new method for reversed phase HPLC determination of thiamine and its major in vivo phosphorylation products, thiamine monophosphate (TMP) and thiamine pyrophosphate (TPP), was developed using tetrabutylammonium hydroxide as the ion-pairing agent. The separation was performed on a Phenomenex Kinetex EVO C18 column with a gradient of a phosphate-buffered aqueous solution of the ion-pair reagent and methanol. The duty cycle for the assay was 13 min and pyrithiamine was successfully used as the internal standard for the first time in a thiamine HPLC measurement protocol. Detection of the fluorescence derivatives of the analytes as well as the IS allowed for lower detection limits in order to support biological applications in cell culture models. The linearity, sensitivity, specificity, accuracy and precision of the method were evaluated and met the requirements specified by the US Food and Drug Administration. The calibration curves proved to be linear and the method was validated over the range from 1.0-4000 nM for both cells and the media where complete recovery of the analytes was also achieved.


Subject(s)
Chromatography, High Pressure Liquid/methods , Thiamine/analysis , Cell Line , Homeostasis , Humans , Thiamine/metabolism
5.
Cancer Chemother Pharmacol ; 73(3): 585-94, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24452394

ABSTRACT

PURPOSE: The dichotomous effect of thiamine supplementation on cancer cell growth is characterized by growth stimulation at low doses and growth suppression at high doses. Unfortunately, how thiamine reduces cancer cell proliferation is currently unknown. Recent focuses on metabolic targets for cancer therapy have exploited the altered regulation of the thiamine-dependent enzyme pyruvate dehydrogenase (PDH). Cancer cells inactivate PDH through phosphorylation by overexpression of pyruvate dehydrogenase kinases (PDKs). Inhibition of PDKs by dichloracetate (DCA) exhibits a growth suppressive effect in many cancers. Recently, it has been shown that the thiamine coenzyme, thiamine pyrophosphate reduces PDK-mediated phosphorylation of PDH. Therefore, the objective of this study was to determine whether high-dose thiamine supplementation reduces cell proliferation through a DCA-like mechanism. METHODS: Cytotoxicity of thiamine and DCA was assessed in SK-N-BE and Panc-1 cancer cell lines. Comparative effects of high-dose thiamine and DCA on PDH phosphorylation were measured by Western blot. The metabolic impact of PDH reactivation was determined by glucose and lactate assays. Changes in the mitochondrial membrane potential, reactive oxygen species (ROS) production, and caspase-3 activation were assessed to characterize the mechanism of action. RESULTS: Thiamine exhibited a lower IC50 value in both cell lines compared with DCA. Both thiamine and DCA reduced the extent of PDH phosphorylation, reduced glucose consumption, lactate production, and mitochondrial membrane potential. High-dose thiamine and DCA did not increase ROS, but increased caspase-3 activity. CONCLUSION: Our findings suggest that high-dose thiamine reduces cancer cell proliferation by a mechanism similar to that described for dichloroacetate.


Subject(s)
Dichloroacetic Acid/administration & dosage , Neuroblastoma/pathology , Pancreatic Neoplasms/pathology , Thiamine/administration & dosage , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Glucose/metabolism , Humans , Lactic Acid/biosynthesis , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Neuroblastoma/drug therapy , Pancreatic Neoplasms/drug therapy , Phosphorylation/drug effects , Pyruvate Dehydrogenase Complex/antagonists & inhibitors , Pyruvate Dehydrogenase Complex/metabolism , Reactive Oxygen Species/metabolism
6.
Cancer Metab ; 1(1): 16, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-24280319

ABSTRACT

The resurgence of interest in cancer metabolism has linked alterations in the regulation and exploitation of metabolic pathways with an anabolic phenotype that increases biomass production for the replication of new daughter cells. To support the increase in the metabolic rate of cancer cells, a coordinated increase in the supply of nutrients, such as glucose and micronutrients functioning as enzyme cofactors is required. The majority of co-enzymes are water-soluble vitamins such as niacin, folic acid, pantothenic acid, pyridoxine, biotin, riboflavin and thiamine (Vitamin B1). Continuous dietary intake of these micronutrients is essential for maintaining normal health. How cancer cells adaptively regulate cellular homeostasis of cofactors and how they can regulate expression and function of metabolic enzymes in cancer is underappreciated. Exploitation of cofactor-dependent metabolic pathways with the advent of anti-folates highlights the potential vulnerabilities and importance of vitamins in cancer biology. Vitamin supplementation products are easily accessible and patients often perceive them as safe and beneficial without full knowledge of their effects. Thus, understanding the significance of enzyme cofactors in cancer cell metabolism will provide for important dietary strategies and new molecular targets to reduce disease progression. Recent studies have demonstrated the significance of thiamine-dependent enzymes in cancer cell metabolism. Therefore, this review discusses the current knowledge in the alterations in thiamine availability, homeostasis, and exploitation of thiamine-dependent pathways by cancer cells.

7.
J Nutr Biochem ; 24(9): 1616-24, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23642734

ABSTRACT

An increased carbon flux and exploitation of metabolic pathways for the rapid generation of biosynthetic precursors is a common phenotype observed in breast cancer. To support this metabolic phenotype, cancer cells adaptively regulate the expression of glycolytic enzymes and nutrient transporters. However, activity of several enzymes involved in glucose metabolism requires an adequate supply of cofactors. In particular, vitamin B1 (thiamine) is utilized as an essential cofactor for metabolic enzymes that intersect at critical junctions within the glycolytic network. Intracellular availability of thiamine is facilitated by the activity of thiamine transporters and thiamine pyrophosphokinase-1 (TPK-1). Therefore, the objective of this study was to establish if the cellular determinants regulating thiamine homeostasis differ between breast cancer and normal breast epithelia. Employing cDNA arrays of breast cancer and normal breast epithelial tissues, SLC19A2, SLC25A19 and TPK-1 were found to be significantly up-regulated. Similarly, up-regulation was also observed in breast cancer cell lines compared to human mammary epithelial cells. Thiamine transport assays and quantitation of intracellular thiamine and thiamine pyrophosphate established a significantly greater extent of thiamine transport and free thiamine levels in breast cancer cell lines compared to human mammary epithelial cells. Overall, these findings demonstrate an adaptive response by breast cancer cells to increase cellular availability of thiamine.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Thiamine/pharmacology , Up-Regulation , Biological Transport , Breast Neoplasms/metabolism , Cell Line, Tumor , Epithelial Cells/metabolism , Female , Gene Expression Profiling , Homeostasis , Humans , MCF-7 Cells , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Thiamin Pyrophosphokinase/genetics , Thiamin Pyrophosphokinase/metabolism
8.
Pharm Res ; 30(4): 1050-64, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23224979

ABSTRACT

PURPOSE: To investigate the role of membrane-associated drug transporters in regulating the intestinal absorption of the HIV-1 protease inhibitor, atazanavir, and assess the potential contribution of these transporters in clinical interactions of atazanavir with other protease inhibitors and tenofovir disoproxil fumarate (TDF). METHODS: Intestinal permeability of atazanavir was investigated in vitro, using the Caco-2 cell line system grown on Transwell inserts, and in situ, by single-pass perfusion of rat intestinal segments, jejunum and ileum, in the absence or presence of standard transporter inhibitors or antiretroviral drugs. RESULTS: Atazanavir accumulation by Caco-2 cells was susceptible to inhibition by P-glycoprotein and organic anion transporting polypeptide (OATP) family inhibitors and several antiretroviral drugs (protease inhibitors, TDF). The secretory flux of atazanavir (basolateral-to-apical Papp) was 11.7-fold higher than its absorptive flux. This efflux ratio was reduced to 1.5-1.7 in the presence of P-glycoprotein inhibitors or ritonavir. P-glycoprotein inhibition also resulted in 1.5-2.5-fold increase in atazanavir absorption in situ. Co-administration of TDF, however, reduced atazanavir intestinal permeability by 13-49%, similar to the effect observed clinically. CONCLUSIONS: Drug transporters such as P-glycoprotein and OATPs regulate intestinal permeability of atazanavir and may contribute to its poor oral bioavailability and drug-drug interactions with other protease inhibitors and TDF.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Anti-HIV Agents/pharmacology , Anti-HIV Agents/pharmacokinetics , Intestinal Absorption , Oligopeptides/pharmacology , Oligopeptides/pharmacokinetics , Organic Anion Transporters/metabolism , Pyridines/pharmacology , Pyridines/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Atazanavir Sulfate , Caco-2 Cells , Drug Interactions , Humans , Intestinal Absorption/drug effects , Intestinal Mucosa/metabolism , Male , Organophosphonates/pharmacology , Permeability/drug effects , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Inhibitors/pharmacology , Tenofovir
9.
Int J Vitam Nutr Res ; 83(3): 188-97, 2013.
Article in English | MEDLINE | ID: mdl-24846908

ABSTRACT

It is well established that thiamine deficiency results in an excess of metabolic intermediates such as lactate and pyruvate, which is likely due to insufficient levels of cofactor for the function of thiamine-dependent enzymes. When in excess, both pyruvate and lactate can increase the stabilization of the hypoxia-inducible factor 1-alpha (HIF-1α) transcription factor, resulting in the trans-activation of HIF-1α regulated genes independent of low oxygen, termed pseudo-hypoxia. Therefore, the resulting dysfunction in cellular metabolism and accumulation of pyruvate and lactate during thiamine deficiency may facilitate a pseudo-hypoxic state. In order to investigate the possibility of a transcriptional relationship between hypoxia and thiamine deficiency, we measured alterations in metabolic intermediates, HIF-1α stabilization, and gene expression. We found an increase in intracellular pyruvate and extracellular lactate levels after thiamine deficiency exposure to the neuroblastoma cell line SK-N-BE. Similar to cells exposed to hypoxia, there was a corresponding increase in HIF-1α stabilization and activation of target gene expression during thiamine deficiency, including glucose transporter-1 (GLUT1), vascular endothelial growth factor (VEGF), and aldolase A. Both hypoxia and thiamine deficiency exposure resulted in an increase in the expression of the thiamine transporter SLC19A3. These results indicate thiamine deficiency induces HIF-1α-mediated gene expression similar to that observed in hypoxic stress, and may provide evidence for a central transcriptional response associated with the clinical manifestations of thiamine deficiency.


Subject(s)
Gene Expression/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Thiamine Deficiency/physiopathology , Cell Hypoxia/physiology , Cell Nucleus/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Lactic Acid/metabolism , Neuroblastoma , Neurons/ultrastructure , Pyruvic Acid/metabolism
10.
J Pharmacol Exp Ther ; 334(3): 1009-22, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20507927

ABSTRACT

Human intestinal epithelium expresses a number of drug efflux and influx transporters that can restrict and/or facilitate intestinal drug uptake during absorption. Organic anion-transporting polypeptide 2B1 (OATP2B1), a multispecific organic anion uptake transporter localized at the brush-border membrane of intestinal epithelial cells, is known to transport many endogenous substrates (e.g., steroid conjugates) and xenobiotics (e.g., statins). At present, limited information is available on the mechanism of HIV protease inhibitor (PIs) intestinal uptake. In this study, we examined the interaction of PIs with the OATP2B1 transport system in Caco-2 cells, an in vitro model of human intestinal epithelium, and Madin-Darby canine kidney II cells stably transfected with OATP2B1. The expression of OATP2B1 transcript and protein was confirmed by reverse transcription-polymerase chain reaction and immunoblot analysis, respectively. Estrone-3-sulfate (E3S) uptake demonstrated biphasic saturation kinetics in Caco-2 cells, with dissociation constants (K(M)) of 6 +/- 2 microM and 1.5 +/- 0.2 mM. Several PIs potently inhibited OATP2B1-mediated transport in Caco-2 cells at clinically relevant IC(50) concentrations for ritonavir (0.93 microM), atazanavir (2.2 microM), lopinavir (1.7 microM), tipranavir (0.77 microM), and nelfinavir (2.2 microM). An inwardly directed proton gradient was identified as the driving force of E3S uptake through NH(4)Cl intracellular acidification studies with a H(+):E3S stoichiometry for OATP2B1 of 1:1. In contrast, although atazanavir and ritonavir uptake by Caco-2 cells was stimulated by low extracellular pH, this process was not mediated by OATP2B1 and was not affected by an outwardly directed H(+) gradient. Because OATP2B1 exhibits an increasing number of drug substrates, including several statins, alterations of its function by PIs could result in clinically significant drug-drug interactions in the intestine.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Organic Anion Transporters/metabolism , Algorithms , Animals , Anti-HIV Agents/pharmacology , Atazanavir Sulfate , Biological Availability , Biological Transport, Active , Blotting, Western , Caco-2 Cells , Cell Line , Dogs , Drug Interactions , Estrone/analogs & derivatives , Estrone/metabolism , Humans , Hydrogen-Ion Concentration , Oligopeptides/pharmacokinetics , Oligopeptides/pharmacology , Organic Anion Transporters/chemistry , Pyridines/pharmacokinetics , Pyridines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Ritonavir/pharmacokinetics , Ritonavir/pharmacology
11.
J Neurosci Res ; 87(4): 1023-36, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18855943

ABSTRACT

A major concern regarding the chronic administration of antiretroviral drugs is the potential for induction of drug efflux transporter expression (i.e., P-glycoprotein, P-gp) at tissue sites that can significantly affect drug distribution and treatment efficacy. Previous data have shown that the inductive effect of human immunodeficiency virus protease inhibitors (PIs) is mediated through the human orphan nuclear receptor, steroid xenobiotic receptor (SXR or hPXR). The objectives of this study were to investigate transport and inductive properties on efflux drug transporters of two PIs, atazanavir and ritonavir, at the blood-brain barrier by using a human brain microvessel endothelial cell line, hCMEC/D3. Transport properties of PIs by the drug efflux transporters P-gp and multidrug resistance protein 1 (MRP1) were assessed by measuring the cellular uptake of (3)H-atazanavir or (3)H-ritonavir in P-gp and MRP1 overexpressing cells as well as hCMEC/D3. Whereas the P-gp inhibitor, PSC833, increased atazanavir and ritonavir accumulation in hCMEC/D3 cells by 2-fold, the MRP inhibitor MK571 had no effect. P-gp, MRP1, and hPXR expression and localization were examined by Western blot analysis and immunogold cytochemistry at the electron microscope level. Treatment of hCMEC/D3 cells for 72 hr with rifampin or SR12813 (two well-established hPXR ligands) or PIs (atazanavir or ritonavir) resulted in an increase in P-gp expression by 1.8-, 6-, and 2-fold, respectively, with no effect observed for MRP1 expression. In hCMEC/D3 cells, cellular accumulation of these PIs appears to be primarily limited by P-gp efflux activity. Long-term exposure of atazanavir or ritonavir to brain microvessel endothelium may result in further limitations in brain drug permeability as a result of the up-regulation of P-gp expression and function.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Endothelium, Vascular/metabolism , HIV Protease Inhibitors/pharmacology , Up-Regulation , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Atazanavir Sulfate , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/blood supply , Brain/drug effects , Brain/metabolism , Cell Line , Cyclosporins/pharmacology , Diphosphonates/pharmacology , Endothelium, Vascular/drug effects , Humans , Microvessels/drug effects , Microvessels/metabolism , Oligopeptides/pharmacology , Propionates/pharmacology , Pyridines/pharmacology , Quinolines/pharmacology , Rifampin/pharmacology , Ritonavir/pharmacology , Tritium/metabolism
12.
Mol Pharm ; 5(4): 643-53, 2008.
Article in English | MEDLINE | ID: mdl-18380467

ABSTRACT

The utilization of surfactants to increase intestinal absorption of drugs is a viable strategy that benefits from increases in drug solubilization and the potential for inhibition of P-glycoprotein (P-gp) mediated efflux. However, the effective concentration range for P-gp inhibition of most surfactants is defined over a narrow concentration range, below the critical micelle concentration (CMC), as a result of significant micelle sequestration of drug. Therefore, the objectives of these studies were to assess if association of P-gp substrates differing in hydrophobicity will impact the effective concentration range for P-gp inhibition by amphiphilic diblock copolymers based on methoxypolyethylene glycol-block-polycaprolatone (MePEG-b-PCL). Comparisons between the micelle association and Caco-2 cellular accumulation were evaluated using two structurally homologous P-gp substrates, the relatively hydrophobic R-6G and the hydrophilic R-123, over concentrations above and below the CMC for MePEG-b-PCL diblock copolymers. An approximately 3.75-fold enhancement of R-123 accumulation occurred with 2 mM MePEG17-b-PCL5, compared to approximately 1.25-fold for R-6G. This decrease in the accumulation enhancement corresponds with the higher R-6G fraction (0.75) associated at 2 mM MePEG17-b-PCL5 compared with R-123 (0.25). Interestingly, R-6G accumulation was enhanced over a very broad range of MePEG17-b-PCL5 concentrations below the CMC. This was in contrast to R-123, which demonstrated no enhancement below the CMC. A similar concentration dependent accumulation profile was seen with other surfactants such as vitamin E TPGS and Cremophor EL and with two other P-gp substrates differing in hydrophobicity, the relatively hydrophobic paclitaxel and hydrophilic doxorubicin. In conclusion, the effective concentration range for surfactant mediated inhibition of P-gp appears to depend on the P-gp substrate hydrophobicity.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Ethylene Oxide/chemistry , Hydrophobic and Hydrophilic Interactions , Lactones/chemistry , Caco-2 Cells , Doxorubicin/chemistry , Humans , Molecular Structure , Paclitaxel/chemistry , Polyethylene Glycols , Surface-Active Agents/chemistry
13.
Free Radic Biol Med ; 33(2): 266-75, 2002 Jul 15.
Article in English | MEDLINE | ID: mdl-12106822

ABSTRACT

The iron chelating hydroxypyridinone deferiprone (CP20, L1) and the clinically approved cardioprotective agent dexrazoxane (ICRF-187) were examined for their ability to protect neonatal rat cardiac myocytes from doxorubicin-induced damage. Doxorubicin is thought to induce oxidative stress on the heart muscle, both through reductive activation to its semiquinone form, and by the production of hydroxyl radicals mediated by its complex with iron. The results of this study showed that both deferiprone and dexrazoxane were able to protect myocytes from doxorubicin-induced lactate dehydrogenase release. Deferiprone quickly and efficiently removed iron(III) from its complex with doxorubicin. In addition, this study also showed that deferiprone rapidly entered myocytes and displaced iron from a fluorescence-quenched trapped intracellular iron-calcein complex, suggesting that in the myocyte, deferiprone should also be able to displace iron from its complex with doxorubicin. It was shown by electron paramagnetic resonance spectroscopy that under hypoxic conditions myocytes were able to reduce doxorubicin to its semiquinone free radical. Deferiprone also greatly reduced hydroxyl radical production by the iron(III)-doxorubicin complex in the xanthine oxidase/xanthine superoxide generating system. Together these results suggest that deferiprone may protect against doxorubicin-induced damage to myocytes by displacing iron bound to doxorubicin, or chelating free or loosely bound iron, thus preventing site-specific iron-based oxygen radical damage.


Subject(s)
Antineoplastic Agents/toxicity , Doxorubicin/toxicity , Iron Chelating Agents/pharmacology , Myocytes, Cardiac/drug effects , Pyridones/pharmacology , Animals , Animals, Newborn , Cell Survival/drug effects , Cells, Cultured , Deferiprone , Electron Spin Resonance Spectroscopy , Fluoresceins/metabolism , Free Radicals/metabolism , Indicators and Reagents/metabolism , Iron/metabolism , L-Lactate Dehydrogenase/metabolism , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Razoxane/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...