Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Int J Mol Sci ; 22(6)2021 Mar 11.
Article in English | MEDLINE | ID: mdl-33799598

ABSTRACT

We sought to develop a cell-based cytotoxicity assay using human hepatocytes, which reflect the effects of drug-metabolizing enzymes on cytotoxicity. In this study, we generated luminescent human hepatoblastoma HepG2 cells using the mouse artificial chromosome vector, in which click beetle luciferase alone or luciferase and major drug-metabolizing enzymes (CYP2C9, CYP2C19, CYP2D6, and CYP3A4) are expressed, and monitored the time-dependent changes of CYP-mediated cytotoxicity expression by bioluminescence measurement. Real-time bioluminescence measurement revealed that compared with CYP-non-expressing cells, the luminescence intensity of CYP-expressing cells rapidly decreased when the cells were treated with low concentrations of aflatoxin B1 or primaquine, which exhibits cytotoxicity in the presence of CYP3A4 or CYP2D6, respectively. Using kinetics data obtained by the real-time bioluminescence measurement, we estimated the time-dependent changes of 50% inhibitory concentration (IC50) values in the aflatoxin B1- and primaquine-treated cell lines. The first IC50 value was detected much earlier and at a lower concentration in primaquine-treated CYP-expressing HepG2 cells than in primaquine-treated CYP-non-expressing cells, and the decrease of IC50 values was much faster in the former than the latter. Thus, we successfully monitored time- and concentration-dependent dynamic changes of CYP-mediated cytotoxicity expression in CYP-expressing luminescent HepG2 cells by means of real-time bioluminescence measurement.


Subject(s)
Aflatoxin B1/toxicity , Founder Effect , Luminescent Measurements/methods , Primaquine/toxicity , Time-Lapse Imaging/methods , Xenobiotics/toxicity , Animals , Cell Line, Tumor , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2C19/metabolism , Cytochrome P-450 CYP2C9/genetics , Cytochrome P-450 CYP2C9/metabolism , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Gene Expression Regulation , Genes, Reporter , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Luciferases/genetics , Luciferases/metabolism , Luminescence , Mice
2.
J Spec Oper Med ; 19(3): 14-16, 2019.
Article in English | MEDLINE | ID: mdl-31539427

ABSTRACT

Presumptive antirelapse therapy (PART) with primaquine for Plasmodium vivax malaria postdeployment is an important component of the US military Force Health Protection plan. While primaquine is well tolerated in the majority of cases, we present a unique case of an active duty Army Ranger without glucose-6-phosphatase dehydrogenase or cytochrome b5 reductase (b5R) deficiencies who developed symptomatic methemoglobinemia while taking PART following a deployment to Afghanistan.


Subject(s)
Methemoglobinemia/chemically induced , Methemoglobinemia/diagnosis , Military Personnel , Primaquine/toxicity , Afghan Campaign 2001- , Delayed Diagnosis , Humans
3.
Chem Biol Interact ; 255: 63-73, 2016 Aug 05.
Article in English | MEDLINE | ID: mdl-26477383

ABSTRACT

The generation of reactive metabolites from therapeutic agents is one of the major mechanisms of drug-induced liver injury (DILI). In order to evaluate metabolism-related toxicity and improve drug efficacy and safety, we generated a battery of HepG2-derived cell lines that express 14 cytochrome P450s (CYPs) (1A1, 1A2, 1B1, 2A6, 2B6, 2C8, 2C9, 2C18, 2C19, 2D6, 2E1, 3A4, 3A5 and 3A7) individually using a lentiviral expression system. The expression/production of a specific CYP in each cell line was confirmed by an increased abundance of the CYP at both mRNA and protein levels. Moreover, the enzymatic activities of representative CYPs in the corresponding cell lines were also measured. Using our CYP-expressed HepG2 cells, the toxicity of three drugs that could induce DILI (amiodarone, chlorpromazine and primaquine) was assessed, and all of them showed altered (increased or decreased) toxicity compared to the toxicity in drug-treated wild-type HepG2 cells. CYP-mediated drug toxicity examined in our cell system is consistent with previous reports, demonstrating the potential of these cells for assessing metabolism-related drug toxicity. This cell system provides a practical in vitro approach for drug metabolism screening and for early detection of drug toxicity. It is also a surrogate enzyme source for the enzymatic characterization of a particular CYP that contributes to drug-induced liver toxicity.


Subject(s)
Amiodarone/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Cytochrome P-450 Enzyme System/metabolism , Liver/drug effects , Antimalarials/toxicity , Antipsychotic Agents/toxicity , Cell Line , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Chlorpromazine/toxicity , Cytochrome P-450 Enzyme Inhibitors/toxicity , Cytochrome P-450 Enzyme System/genetics , Drug Evaluation, Preclinical/methods , Gene Expression , Hep G2 Cells , Humans , Liver/metabolism , Liver/pathology , Primaquine/toxicity , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics
4.
Antimicrob Agents Chemother ; 58(8): 4737-44, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24913163

ABSTRACT

Hematotoxicity in individuals genetically deficient in glucose-6-phosphate dehydrogenase (G6PD) activity is the major limitation of primaquine (PQ), the only antimalarial drug in clinical use for treatment of relapsing Plasmodium vivax malaria. PQ is currently clinically used in its racemic form. A scalable procedure was developed to resolve racemic PQ, thus providing pure enantiomers for the first time for detailed preclinical evaluation and potentially for clinical use. These enantiomers were compared for antiparasitic activity using several mouse models and also for general and hematological toxicities in mice and dogs. (+)-(S)-PQ showed better suppressive and causal prophylactic activity than (-)-(R)-PQ in mice infected with Plasmodium berghei. Similarly, (+)-(S)-PQ was a more potent suppressive agent than (-)-(R)-PQ in a mouse model of Pneumocystis carinii pneumonia. However, at higher doses, (+)-(S)-PQ also showed more systemic toxicity for mice. In beagle dogs, (+)-(S)-PQ caused more methemoglobinemia and was toxic at 5 mg/kg of body weight/day given orally for 3 days, while (-)-(R)-PQ was well tolerated. In a novel mouse model of hemolytic anemia associated with human G6PD deficiency, it was also demonstrated that (-)-(R)-PQ was less hemolytic than (+)-(S)-PQ for the G6PD-deficient human red cells engrafted in the NOD-SCID mice. All these data suggest that while (+)-(S)-PQ shows greater potency in terms of antiparasitic efficacy in rodents, it is also more hematotoxic than (-)-(R)-PQ in mice and dogs. Activity and toxicity differences of PQ enantiomers in different species can be attributed to their different pharmacokinetic and metabolic profiles. Taken together, these studies suggest that (-)-(R)-PQ may have a better safety margin than the racemate in human.


Subject(s)
Antimalarials/pharmacokinetics , Hemolysis/drug effects , Malaria/drug therapy , Pneumonia, Pneumocystis/drug therapy , Primaquine/pharmacokinetics , Animals , Antimalarials/isolation & purification , Antimalarials/toxicity , Dogs , Erythrocyte Transfusion , Erythrocytes/drug effects , Erythrocytes/parasitology , Female , Glucosephosphate Dehydrogenase Deficiency/metabolism , Humans , Lethal Dose 50 , Malaria/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Mice, Inbred NOD , Mice, SCID , Plasmodium berghei/drug effects , Plasmodium berghei/physiology , Pneumocystis carinii/drug effects , Pneumocystis carinii/physiology , Pneumonia, Pneumocystis/microbiology , Primaquine/isolation & purification , Primaquine/toxicity , Stereoisomerism , Transplantation, Heterologous
5.
Malar J ; 13: 148, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24742291

ABSTRACT

BACKGROUND: Primaquine, the only available drug effective against Plasmodium falciparum sexual stages, induces also a dose-dependent haemolysis, especially in glucose-6-phosphate dehydrogenase deficient (G6PDd) individuals. Therefore, it is important to determine the prevalence of this deficiency in areas that would potentially benefit from its use. The prevalence of G6PD deficiency by genotype and enzyme activity was determined in healthy school children in The Gambia. METHODS: Blood samples from primary school children collected during a dry season malaria survey were screened for G6PDd and malaria infection. Genotypes for allele mutations reported in the country; 376, 202A-, 968A- and 542 were analysed while enzyme activity (phenotype) was assayed using a semi-quantitative commercial test kit. Enzyme activity values were fitted in a finite mixture model to determine the distribution and calculate a cut-off for deficiency. The association between genotype and phenotype for boys and girls as well as the association between mutant genotype and deficient phenotype was analysed. RESULTS: Samples from 1,437 children; 51% boys were analysed. The prevalence of P. falciparum malaria infection was 14%. The prevalence of the 202A-, 968 and 542 mutations was 1.8%, 2.1% and 1.0%, respectively, and higher in boys than in girls. The prevalence of G6PDd phenotype was 6.4% (92/1,437), 7.8% (57/728) in boys and 4.9% (35/709) in girls with significantly higher odds in the former (OR 1.64, 95% CI 1.05, 2.53, p = 0.026). The deficient phenotype was associated with reduced odds of malaria infection (OR 0.77, 95% CI 0.36, 1.62, p = 0.49). CONCLUSIONS: There is a weak association between genotype and phenotype estimates of G6PDd prevalence. The phenotype expression of deficiency represents combinations of mutant alleles rather than specific mutations. Genotype studies in individuals with a deficient phenotype would help identify alleles responsible for haemolysis.


Subject(s)
Antimalarials/toxicity , Genotype , Glucosephosphate Dehydrogenase Deficiency/epidemiology , Glucosephosphate Dehydrogenase/genetics , Primaquine/toxicity , Adolescent , Alleles , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Female , Gambia/epidemiology , Glucosephosphate Dehydrogenase/metabolism , Hemolysis , Humans , Malaria, Falciparum/epidemiology , Male , Plasmodium falciparum/isolation & purification , Polymerase Chain Reaction , Prevalence
6.
Int J Biol Macromol ; 62: 18-24, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23973494

ABSTRACT

Primaquine (PQ) shows activity against the late hepatic stages and latent tissue forms of Plasmodium vivax and Plasmodium ovale. However, liver targeted PQ delivery may be useful to minimize the dose-limiting blood toxicities and side-effects of PQ. The prime objective of this study was the preparation of PQ loaded chitosan nanoparticles (PQ-CS-NPs) in order to enhance drug tolerance and to reduce dose frequency. The morphological analysis of the chitosan NPs displayed particle size in the range 287-686 nm, polydispersity index in the range 0.338-0.430 and zeta potential between 9.21 and 22.80 mV which indicated good stability. PQ-CS-NPs exhibited EE and LC as 64.28 ± 1.85% and 33.18 ± 0.975%, respectively. The in vitro drug release (Batch C7) was 97.80 ± 0.65% after 24 h. After intravenous injection of PQ-CS-NPs in mice, the lethal dose of the PQ significantly reduced when compared to that of free PQ solution.


Subject(s)
Chitosan/chemistry , Drug Carriers/chemistry , Nanoparticles/chemistry , Primaquine/chemistry , Primaquine/toxicity , Toxicity Tests , Animals , Drug Liberation , Drug Stability , Liver/cytology , Liver/drug effects , Liver/metabolism , Mice , Particle Size , Primaquine/metabolism
7.
J Basic Clin Physiol Pharmacol ; 24(2): 105-14, 2013.
Article in English | MEDLINE | ID: mdl-23412873

ABSTRACT

BACKGROUND: Primaquine (Pq) metabolic products are responsible for drug-associated hemotoxicity and limit primaquine usage. METHODS: Methemoglobin (MetHb)-Pq molecular modeling was used to identify the Pq binding pocket. UPLC, mass spectrometry, and other indirect analytical methods were used to predict the metabolite. MetHb generation, development of oxidative stress, inhibition of antioxidant enzymes, and scanning electron microscope (SEM) were used to characterize the hemotoxic potentials of oxidized Pq (Pqox). RESULTS: MetHb binded Pq at the heme site with KD =6.4 µM as evidenced by a difference spectroscopy study. MetHb oxidized Pq through a single e-transfer mechanism to form Pqox. The analysis of Pq from MetHb-H2O2 peroxidase reaction mixture gave peaks at m/z 300.53 and m/z 243.42, corresponding to the hydroxyl and desamino derivative of Pq, respectively. Similar peaks were absent in Pq or Pq incubated with H2O2 in the same buffer system. A robust increase in MetHb formation, reactive oxygen species generation, and inhibition of antioxidant enzymes were found in red blood cells (RBCs) exposed to Pqox compared with a parent drug molecule. The RBC membrane exhibited visible damages to plasma membrane (holes) as evidenced by SEM analysis of Pqox-exposed RBCs. CONCLUSIONS: The MetHb-H2O2 system transforms quiescent parent drug molecule to a highly reactive oxidative form to exhibit severe hemolysis. MetHb-H2O2-mediated Pq hemolytic potentiation that is sensitive to spin trap indicates the role of Pq* radical or other single e-species in the process. The result suggests that MetHb incites the molecular property of the Pq and peroxidase inhibitors can be explored to control drug-associated toxicity.


Subject(s)
Antimalarials/toxicity , Methemoglobin/chemistry , Primaquine/toxicity , Antioxidants/metabolism , Catalase/metabolism , Chromatography, High Pressure Liquid , Erythrocyte Membrane/drug effects , Erythrocytes/drug effects , Glutathione Transferase/metabolism , Hematocrit , Hemolysis/drug effects , Humans , Microscopy, Electron, Scanning , Models, Molecular , Osmotic Pressure , Oxidation-Reduction , Oxidative Stress/drug effects , Protein Binding , Spectrum Analysis , Superoxide Dismutase/metabolism
8.
PLoS Med ; 9(11): e1001339, 2012.
Article in English | MEDLINE | ID: mdl-23152723

ABSTRACT

BACKGROUND: Primaquine is a key drug for malaria elimination. In addition to being the only drug active against the dormant relapsing forms of Plasmodium vivax, primaquine is the sole effective treatment of infectious P. falciparum gametocytes, and may interrupt transmission and help contain the spread of artemisinin resistance. However, primaquine can trigger haemolysis in patients with a deficiency in glucose-6-phosphate dehydrogenase (G6PDd). Poor information is available about the distribution of individuals at risk of primaquine-induced haemolysis. We present a continuous evidence-based prevalence map of G6PDd and estimates of affected populations, together with a national index of relative haemolytic risk. METHODS AND FINDINGS: Representative community surveys of phenotypic G6PDd prevalence were identified for 1,734 spatially unique sites. These surveys formed the evidence-base for a Bayesian geostatistical model adapted to the gene's X-linked inheritance, which predicted a G6PDd allele frequency map across malaria endemic countries (MECs) and generated population-weighted estimates of affected populations. Highest median prevalence (peaking at 32.5%) was predicted across sub-Saharan Africa and the Arabian Peninsula. Although G6PDd prevalence was generally lower across central and southeast Asia, rarely exceeding 20%, the majority of G6PDd individuals (67.5% median estimate) were from Asian countries. We estimated a G6PDd allele frequency of 8.0% (interquartile range: 7.4-8.8) across MECs, and 5.3% (4.4-6.7) within malaria-eliminating countries. The reliability of the map is contingent on the underlying data informing the model; population heterogeneity can only be represented by the available surveys, and important weaknesses exist in the map across data-sparse regions. Uncertainty metrics are used to quantify some aspects of these limitations in the map. Finally, we assembled a database of G6PDd variant occurrences to inform a national-level index of relative G6PDd haemolytic risk. Asian countries, where variants were most severe, had the highest relative risks from G6PDd. CONCLUSIONS: G6PDd is widespread and spatially heterogeneous across most MECs where primaquine would be valuable for malaria control and elimination. The maps and population estimates presented here reflect potential risk of primaquine-associated harm. In the absence of non-toxic alternatives to primaquine, these results represent additional evidence to help inform safe use of this valuable, yet dangerous, component of the malaria-elimination toolkit. Please see later in the article for the Editors' Summary.


Subject(s)
Antimalarials/toxicity , Gene Frequency , Geography, Medical/methods , Glucosephosphate Dehydrogenase Deficiency/epidemiology , Glucosephosphate Dehydrogenase/genetics , Hemolysis , Primaquine/toxicity , Bayes Theorem , Female , Geographic Mapping , Glucosephosphate Dehydrogenase/metabolism , Humans , Malaria/epidemiology , Male , Models, Genetic , Prevalence , Risk Assessment , Sex Distribution
9.
Toxicology ; 294(1): 54-60, 2012 Mar 29.
Article in English | MEDLINE | ID: mdl-22330256

ABSTRACT

Therapeutic utility of primaquine, an 8-aminoquinoline antimalarial drug, has been limited due to its hemolytic toxicity in population with glucose 6-phosphate dehydrogenase deficiency. Recent investigations at our lab have shown that the metabolites generated through cytochrome P(450)-dependent metabolic reactions are responsible for hemotoxic effects of primaquine, which could be monitored with accumulation of methemoglobin and increased oxidative stress. The molecular markers for succeeding cascade of events associated with early clearance of the erythrocytes from the circulation were evaluated for understanding the mechanism for hemolytic toxicity of primaquine. Primaquine alone though did not induce noticeable methemoglobin accumulation, but produced significant oxidative stress, which was higher in G6PD-deficient than in normal erythrocytes. Primaquine, presumably through redox active hemotoxic metabolites generated in situ in human liver microsomal metabolism-linked assay, induced a dose-dependent methemoglobin accumulation and oxidative stress, which were almost similar in normal and G6PD-deficient erythrocytes. Primaquine alone or in presence of pooled human liver microsomes neither produced significant effect on intraerythrocytic calcium levels nor affected the phosphatidyl serine asymmetry of the normal and G6PD-deficient human erythrocytes as monitored flowcytometrically with Annexin V binding assay. The studies suggest that eryptosis mechanisms are not involved in accelerated removal of erythrocytes due to hemolytic toxicity of primaquine.


Subject(s)
Erythrocytes/drug effects , Glucosephosphate Dehydrogenase Deficiency/blood , Hemolysis/drug effects , Primaquine/toxicity , Annexins/metabolism , Calcium/blood , Dose-Response Relationship, Drug , Erythrocytes/enzymology , Glucosephosphate Dehydrogenase/blood , Glucosephosphate Dehydrogenase Deficiency/metabolism , Humans , Methemoglobin/analysis , Methemoglobin/biosynthesis , Methemoglobin/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/blood
10.
Chem Res Toxicol ; 24(9): 1476-85, 2011 Sep 19.
Article in English | MEDLINE | ID: mdl-21699254

ABSTRACT

The electron affinities (EA) of the 8-aminoquinoline antimalarial drug primaquine and several of its metabolites were studied using the density functional theory method. We first considered six substituents at the 5-position, -CH(3), -OH, -OCH(3), -Ph, -OPh, and -CHO. We found that in the gas phase the adiabatic EAs are similar to that of the parent primaquine for the -CH(3), -OH, and -OCH(3) substituents. In contrast, the -Ph, -OPh, and -CHO substituents all markedly increase the adiabatic EA. However, only the -CHO substituted compound is predicted to form a stable covalently bound radical anion in the gas phase due to its significant positive vertical EA relative to that of the parent primaquine. In addition, when the 8-position is substituted by the N-hydroxyl group or a quinone-imine structure is formed, the electron capture ability is significantly increased. In aqueous solution, all these molecules have significantly larger adiabatic EAs than in the gas phase. In addition, all of the vertical EAs are positive in aqueous solution. The implications of these findings for contributing to our mechanistic understanding of the red cell toxicity of 8-aminoquinoline compounds are further discussed.


Subject(s)
Aminoquinolines/chemistry , Antimalarials/chemistry , Free Radicals/chemistry , Primaquine/analogs & derivatives , Primaquine/chemistry , Aminoquinolines/toxicity , Anions/chemistry , Anions/toxicity , Antimalarials/toxicity , Electrons , Free Radicals/toxicity , Models, Molecular , Primaquine/toxicity , Quantum Theory
11.
Toxicol Appl Pharmacol ; 241(1): 14-22, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19616568

ABSTRACT

Primaquine, an 8-aminoquinoline, is the drug of choice for radical cure of relapsing malaria. Use of primaquine is limited due to its hemotoxicity, particularly in populations with glucose-6-phosphate dehydrogenase deficiency [G6PD(-)]. Biotransformation appears to be central to the anti-infective and hematological toxicities of primaquine, but the mechanisms are still not well understood. Metabolic studies with primaquine have been hampered due to the reactive nature of potential hemotoxic metabolites. An in vitro metabolism-linked hemotoxicity assay has been developed. Co-incubation of the drug with normal or G6PD(-) erythrocytes, microsomes or recombinant cytochrome P(450) (CYP) isoforms has allowed in situ generation of potential hemotoxic metabolite(s), which interact with the erythrocytes to generate hemotoxicity. Methemoglobin formation, real-time generation of reactive oxygen intermediates (ROIs) and depletion of reactive thiols were monitored as multiple biochemical end points for hemotoxicity. Primaquine alone did not produce any hemotoxicity, while a robust increase was observed in methemoglobin formation and generation of ROIs by primaquine in the presence of human or mouse liver microsomes. Multiple CYP isoforms (CYP2E1, CYP2B6, CYP1A2, CYP2D6 and CYP3A4) variably contributed to the hemotoxicity of primaquine. This was further confirmed by significant inhibition of primaquine hemotoxicity by the selective CYP inhibitors, namely thiotepa (CYP2B6), fluoxetine (CYP2D6) and troleandomycin (CYP3A4). Primaquine caused similar methemoglobin formation in G6PD(-) and normal human erythrocytes. However, G6PD(-) erythrocytes suffered higher oxidative stress and depletion of thiols than normal erythrocytes due to primaquine toxicity. The results provide significant insights regarding CYP isoforms contributing to hemotoxicity and may be useful in controlling toxicity of primaquine to increase its therapeutic utility.


Subject(s)
Antimalarials/toxicity , Cytochrome P-450 Enzyme System/metabolism , Erythrocytes/drug effects , Microsomes, Liver/drug effects , Primaquine/toxicity , Animals , Antimalarials/metabolism , Enzyme Inhibitors/pharmacology , Female , Fluoxetine/pharmacology , Glucosephosphate Dehydrogenase/metabolism , Humans , Isoenzymes/metabolism , Male , Methemoglobin/drug effects , Methemoglobin/metabolism , Mice , Mice, Inbred ICR , Microsomes, Liver/metabolism , Oxidative Stress/drug effects , Primaquine/metabolism , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/metabolism , Thiotepa/pharmacology , Troleandomycin/pharmacology
12.
Toxicology ; 261(1-2): 9-18, 2009 Jun 30.
Article in English | MEDLINE | ID: mdl-19447539

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), a known disruptor of B-cell differentiation and a ligand for the aryl hydrocarbon receptor (AhR), induces binding of the AhR to dioxin responsive elements (DRE) in sensitive genes. The Ig heavy chain (IgH) gene is a sensitive target of TCDD and may be transcriptionally inhibited by TCDD through inhibition of the 3'IgH transcriptional regulatory region (3'IgHRR). While the 3'IgHRR contains binding sites for several transcription factors, two DRE motifs were also identified which may be responsible for TCDD-induced inhibition of 3'IgHRR activation and may implicate the AhR as an important regulator of IgH expression. The objectives of the present study were to determine if 3'IgHRR modulation is limited to TCDD or if structurally diverse chemicals (AhR ligands and non-AhR ligands) from environmental, industrial, dietary or pharmaceutical origin are also capable of modulating the 3'IgHRR and to verify a correlation between effects on a stable 3'IgHRR reporter and the endogenous IgH protein. Utilizing a CH12.LX mouse B-cell line that stably expresses a 3'IgHRR-regulated transgene, we identified an inhibition of both 3'IgHRR activation and IgH protein expression by the non-dioxin AhR activators indolo(3,2-b)carbazole, primaquine, carbaryl, and omeprazole which followed a rank order potency for AhR activation supporting a role of the AhR in the transcriptional regulation of the 3'IgHRR and IgH expression. However, modulation of the 3'IgHRR and IgH expression was not limited to AhR activators or to suppressive effects. Hydrogen peroxide and terbutaline had an activating effect and benzyl isothiocyanate was inhibitory. These chemicals are not known to influence the AhR signaling pathway but have been previously shown to modulate humoral immunity and/or transcription factors that regulate the 3'IgHRR. Taken together these results implicate the 3'IgHRR as a sensitive immunological target and are the first to identify altered 3'IgHRR activation by a diverse range of chemicals.


Subject(s)
Immunoglobulin Heavy Chains/drug effects , Receptors, Aryl Hydrocarbon/drug effects , Regulatory Sequences, Nucleic Acid/drug effects , Transcription, Genetic/drug effects , Xenobiotics/toxicity , Animals , Basic Helix-Loop-Helix Transcription Factors , Carbaryl/toxicity , Carbazoles/toxicity , Cell Line, Tumor , Dose-Response Relationship, Drug , Genes, Reporter , Immunoglobulin Heavy Chains/genetics , Immunoglobulin gamma-Chains/genetics , Ligands , Mice , Molecular Structure , Omeprazole/toxicity , Polychlorinated Dibenzodioxins/toxicity , Primaquine/toxicity , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , Transfection , Xenobiotics/chemistry
13.
J Med Toxicol ; 4(3): 157-66, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18821488

ABSTRACT

INTRODUCTION: Many substances that form methemoglobin (MHb) effectively counter cyanide (CN) toxicity. Although MHb formers are generally applied as treatments for CN poisoning, it has been proposed that a stable, long-acting MHb former could serve as a CN pretreatment. Using this rationale, the 8-aminoquinoline WR242511, a potent long-lasting MHb former in rodents and beagle dogs, was studied in the rhesus monkey for advanced development as a potential CN pretreatment. METHODS: In this study, WR242511 was administered intravenously (IV) in 2 female and 4 male rhesus monkeys in doses of 3.5 and/or 7.0 mg/kg; a single male also received WR242511 orally (PO) at 7.0 mg/kg. Health status and MHb levels were monitored following exposure. RESULTS: The selected doses of WR242511, which produced significant methemoglobinemia in beagle dogs in earlier studies conducted elsewhere, produced very little MHb (mean < 2.0%) in the rhesus monkey. Furthermore, transient hemoglobinuria was noted approximately 60 minutes postinjection of WR242511 (3.5 or 7.0 mg/kg), and 2 lethalities occurred (one IV and one PO) following the 7.0 mg/kg dose. Myoglobinuria was also observed following the 7.0 mg/kg dose. Histopathology analyses in the 2 animals that died revealed liver and kidney toxicity, with greater severity in the orally-treated animal. CONCLUSIONS: These data demonstrate direct and/or indirect drug-induced toxicity. It is concluded that WR242511 should not be pursued as a pretreatment for CN poisoning unless the anti-CN characteristics of this compound can be successfully dissociated from those producing undesirable toxicity.


Subject(s)
Antimalarials/toxicity , Primaquine/analogs & derivatives , Animals , Female , Hemoglobinuria/chemically induced , Immobilization , Kidney/pathology , Liver/pathology , Lung/pathology , Macaca mulatta , Male , Methemoglobin/metabolism , Myoglobinuria/chemically induced , Primaquine/toxicity , Solvents
14.
Basic Clin Pharmacol Toxicol ; 103(6): 522-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18684221

ABSTRACT

Therapeutic agents derived from 8-aminoquinoline possess potent activity against hepatic stages of plasmodia. Bulaquine (CDRI 80/53), an enamine analogue of primaquine and a relatively new derivative of 8-aminioquinoline, synthesized at the Central Drug Research Institute, Lucknow, India, has shown promising activity against hypnozoites of Plasmodium vivax and Plasmodium ovale. Moreover, it has been found to be three to four times safer than primaquine in pre-clinical studies. In this study, global gene profiling using 22,827 probes was carried out in the livers of male Swiss mice to identify affected genes and cellular pathways at 6, 12 and 24 hr after a single oral dose of bulaquine (40 mg/kg). Present gene expression analysis revealed perturbation in 11 probes (P < 0.01 and 2-fold), including those corresponding to protein synthesis, cell division, protein ubiquitination, transcription regulation and steroid biosynthesis. Large numbers of probes (>100) corresponding to transcription, protein biosynthesis and intracellular signalling showed >2-fold differential expression at one of the time-points. Furthermore, 60 Gene Ontology terms were affected significantly (z score > 2). Conversely, serum biochemistry and histological evaluation of hepatic tissue showed no signs of stress. These gene expression alterations provide the first report of early hepatic response after an acute dose of bulaquine in mice liver; however, absence of traditional markers of hepatic stress might suggest a general hepatic response inherent in these transcriptional changes. Interestingly, the total number of affected probes was less as compared to that previously reported for primaquine.


Subject(s)
Antimalarials/toxicity , Liver/drug effects , Primaquine/analogs & derivatives , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Dose-Response Relationship, Drug , Gene Expression Profiling , Liver/metabolism , Male , Mice , Oligonucleotide Array Sequence Analysis , Oxidative Stress/drug effects , Primaquine/toxicity , RNA, Messenger/metabolism
15.
Photochem Photobiol ; 83(6): 1415-27, 2007.
Article in English | MEDLINE | ID: mdl-18028216

ABSTRACT

This article describes the results of a coupled photophysical and photobiological study aimed at understanding the phototoxicity mechanism of the antimalarial drugs amodiaquine (AQ), primaquine (PQ) and chloroquine (CQ). Photophysical experiments were carried out in aqueous solutions by steady-state and time-resolved spectrometric techniques to obtain information on the different decay pathways of the excited states of the drugs and on the transient species formed upon laser irradiation. The results showed that all three drugs possess very low fluorescence quantum yields (10(-2)-10(-4)). Laser flash photolysis experiments proved the occurrence of photoionization processes leading to the formation of a radical cation in all three systems. In the case of AQ the lowest triplet state was also detected. Together with the photophysical properties the photobiological properties of the antimalarial drugs were investigated under UV irradiation, on various biological targets through a series of in vitro assays. Phototoxicity on mouse 3T3 fibroblast and human keratinocyte cell lines NCTC-2544 was detected for PQ and CQ but not for AQ. In particular, PQ- and CQ-induced apoptosis was revealed by the externalization of phosphatidylserine. Furthermore, upon UV irradiation, the drugs caused significant variations of the mitochondrial potential (Deltapsi(mt)) measured by flow cytometry. The photodamages produced by the drugs were also evaluated on proteins, lipids and DNA. The combined approaches were useful in understanding the mechanism of phototoxicity induced by these antimalarial drugs.


Subject(s)
Amodiaquine/chemistry , Chloroquine/chemistry , Primaquine/chemistry , Amodiaquine/toxicity , Animals , Antimalarials , Cell Line , Cell Survival/drug effects , Cell Survival/radiation effects , Chloroquine/toxicity , DNA/genetics , DNA Damage , Humans , Lipid Peroxidation/drug effects , Lipid Peroxidation/radiation effects , Lysosomes/drug effects , Lysosomes/radiation effects , Mice , Mitochondria/drug effects , Mitochondria/radiation effects , Molecular Structure , Photobiology , Photochemistry , Primaquine/toxicity , Reactive Oxygen Species/metabolism , Salmon , Spectrophotometry
16.
Toxicology ; 239(1-2): 96-107, 2007 Sep 24.
Article in English | MEDLINE | ID: mdl-17686563

ABSTRACT

Primaquine (PQ), a clinically important derivative of 8-aminoquinoline used against the hepatic stages (hypnozoites) of Plasmodium vivax and Plasmodium ovale, was studied to evaluate and compare between mRNA expression, and biochemical and histological parameters of hepatic stress in adult Swiss mice (Mus musculus). Following single oral dose of PQ (40 mg/kg, bw), alanine aminotransferase (ALT) and aspartate aminotransferase (AST) along with hematoxylin and eosin stained liver sections did not show any signs of hepatic stress at 6, 12 and 24 h except for ALT activity at 6h. However, analysis at RNA transcript level revealed consistent and significant deregulation (p<0.01 and two-fold) of 16 probes corresponding to important cellular processes such as protein transportation, transcription regulation, intracellular signaling, protein synthesis, hematopoiesis, cell adhesion and cell proliferation. Pathway analysis identified large number of affected genes corresponding to 40 Gene Ontology terms having a z score greater than 2. These results indicate that PQ at high doses may affect gene expression in liver and may produce undesirable outcomes if consumed for longer durations.


Subject(s)
Antimalarials/toxicity , Gene Expression/drug effects , Liver/drug effects , Oligonucleotide Array Sequence Analysis/methods , Primaquine/toxicity , Toxicogenetics/methods , Administration, Oral , Alanine Transaminase/blood , Animals , Animals, Outbred Strains , Aspartate Aminotransferases/blood , Gene Expression Profiling , Liver/metabolism , Liver/pathology , Male , Mice , Oxidative Stress/drug effects , Oxidative Stress/genetics , RNA, Messenger/metabolism
17.
J Pharmacol Exp Ther ; 315(3): 980-6, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16099929

ABSTRACT

Primaquine-induced hemolytic anemia is known to result from premature sequestration of damaged (but intact) erythrocytes by the spleen. We have shown previously that a phenolic metabolite, 5-hydroxyprimaquine (5-HPQ), is a direct-acting hemolytic agent in rats, suggesting that 5-HPQ is a mediator of the hemolytic response to primaquine. To investigate the fate of erythrocytes in vivo after in vitro exposure to 5-HPQ, rat (51)Cr-labeled erythrocytes were incubated with hemolytic concentrations of 5-HPQ and then readministered intravenously to rats. The time course of loss of radioactivity from blood and uptake into the spleen and liver was measured. In rats given 5-HPQ-treated erythrocytes, an increased rate of removal of radioactivity from the circulation was observed as compared with the vehicle control. The loss of blood radioactivity was accompanied by a corresponding increase in radioactivity appearing in the spleen but not in the liver. When rats were pretreated with clodronate-loaded liposomes to deplete splenic macrophages, there was a decreased rate of removal of radioactivity from the circulation and a markedly diminished uptake into the spleen. A role for phagocytic removal of 5-HPQ-treated red cells was confirmed in vitro using the J774A.1 macrophage cell line. Furthermore, depletion of red cell GSH with diethyl maleate significantly enhanced in vitro phagocytosis of 5-HPQ-treated red cells. The data indicate that splenic macrophages are responsible for removing 5-HPQ-treated red cells and support the postulate that this metabolite is a contributor to the hemolytic anemia induced after administration of the parent compound.


Subject(s)
Anemia, Hemolytic/chemically induced , Erythrocytes/drug effects , Macrophages/drug effects , Primaquine/toxicity , Spleen/metabolism , Anemia, Hemolytic/pathology , Animals , Cell Line , Erythrocytes/metabolism , Macrophages/cytology , Macrophages/metabolism , Male , Phagocytosis/drug effects , Primaquine/analogs & derivatives , Primaquine/chemistry , Rats , Rats, Sprague-Dawley
18.
J Pharmacol Exp Ther ; 314(2): 838-45, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15840764

ABSTRACT

Primaquine-induced hemolytic anemia is a toxic side effect that is due to premature splenic sequestration of intact erythrocytes. Previous studies have suggested that a phenolic metabolite, 5-hydroxyprimaquine (5-HPQ), mediates primaquine hemotoxicity by generating reactive oxygen species (ROS) within erythrocytes that overwhelm antioxidant defenses. However, the nature of the oxidative stress is not understood, and the molecular targets, whether protein and/or lipid, are unknown. To investigate the mechanism underlying the hemolytic activity of 5-HPQ, we have examined the effect of hemolytic concentrations of 5-HPQ on ROS formation within rat erythrocytes using the cellular ROS probe, 2',7'-dichlorodihydrofluoresein diacetate. In addition, we examined the effect of 5-HPQ on membrane lipids and cytoskeletal proteins. The data indicate that 5-HPQ causes a prolonged, concentration-dependent generation of ROS within erythrocytes. Interestingly, 5-HPQ-generated ROS was not associated with the onset of lipid peroxidation or an alteration in phosphatidylserine asymmetry. Instead, 5-HPQ induced oxidative injury to the erythrocyte cytoskeleton, as evidenced by changes in the normal electrophoretic pattern of membrane ghost proteins. Immunoblotting with an anti-hemoglobin antibody revealed that these changes were due primarily to the formation of disulfide-linked hemoglobin-skeletal protein adducts. The data suggest that cytoskeletal protein damage, rather than membrane lipid peroxidation or loss of phosphatidylserine asymmetry, underlies the process of removal of erythrocytes exposed to 5-HPQ.


Subject(s)
Anemia, Hemolytic/chemically induced , Antimalarials/toxicity , Cell Membrane/metabolism , Cytoskeletal Proteins/metabolism , Lipid Peroxidation/drug effects , Primaquine/analogs & derivatives , Animals , Calcium/pharmacology , Cell Membrane/drug effects , Dithiothreitol/pharmacology , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , Erythrocyte Membrane/drug effects , Erythrocyte Membrane/metabolism , Erythrocytes/metabolism , Ethylmaleimide/pharmacology , In Vitro Techniques , Ionophores/pharmacology , Lipid Bilayers , Male , Phosphatidylserines/blood , Primaquine/toxicity , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/blood , Spleen/metabolism
19.
J Clin Pharmacol ; 45(3): 346-51, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15703369

ABSTRACT

Hypersensitivity adverse drug reactions are much more common among patients with acquired immunodeficiency syndrome (AIDS) than in the general population. High rates of hypersensitivity reactions to clindamycin have been noted. To investigate the role of reactive metabolites in these reactions, the authors studied toxicity of clindamycin and sulphamethoxazole (SMX) and their metabolites in uninfected and human immunodeficiency virus (HIV)-infected MOLT3 cells. Infected and uninfected cells were incubated with clindamycin or sulphamethoxazole hydroxylamine in increasing concentrations; reactive metabolites were generated by coincubation of cells and drug with murine microsomes and a microsomal activating system. Over a concentration range of 0 to 400 microM SMX-HA, there was a significant concentration-dependent increase in cell death in HIV-infected compared to uninfected cells (28%+/-3% vs 8%+/-5% at 400 microM, P < .05). In contrast, coincubation of cells with clindamycin, microsomes, and a microsomal activating system, as well as combinations of primaquine or pyrimethamine, was not associated with an increase in cell death among infected compared to uninfected cells. No concentration-toxicity was demonstrated. These data support the role of reactive metabolites in adverse drug reactions to sulfonamides during HIV infection, whereas alternate mechanism(s) may be responsible for increased rates of adverse drug reactions to clindamycin among patients with AIDS.


Subject(s)
Anti-Infective Agents/toxicity , Cell Survival/drug effects , Clindamycin/toxicity , HIV/drug effects , Sulfamethoxazole/analogs & derivatives , T-Lymphocytes/drug effects , Animals , Biotransformation , Cell Line , Clindamycin/metabolism , Coculture Techniques , Drug Combinations , Drug Hypersensitivity/etiology , HIV/pathogenicity , Mice , Microsomes , Primaquine/toxicity , Pyrimethamine/toxicity , Sulfamethoxazole/metabolism , Sulfamethoxazole/toxicity , T-Lymphocytes/virology
20.
Drug Chem Toxicol ; 27(1): 41-53, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15038247

ABSTRACT

Effects of in vivo treatment with the three antimalarials chloroquine, primaquine and quinine on rat liver mitochondrial energy transduction functions were examined. Treatment with all the three antimalarials decreased the state 3 and state 4 respiration rates drastically. The extent of inhibition was higher with pyruvate + malate as the substrate than with glutamate. The antimalarials also acted as uncouplers. The uncoupling effect was seen on site II and site III of phosphorylation; site I was unaffected. As a consequence the ADP phosphorylation rates also decreased significantly. Following antimalarials treatment the basal and Mg2+ stimulated ATPase activities increased while the DNP-stimulated ATPase activity was reduced by half. Treatment with chloroquine resulted in decreased contents of cytochromes aa3 and b; primaquine and quinine treatments increased the contents of the two cytochromes in 14 day treatment groups.


Subject(s)
Antimalarials/toxicity , Mitochondria, Liver/drug effects , Adenosine Triphosphatases/drug effects , Adenosine Triphosphatases/metabolism , Animals , Chloroquine/toxicity , Cytochromes/classification , Cytochromes/drug effects , Cytochromes/metabolism , Energy Metabolism/drug effects , Male , Mitochondria, Liver/enzymology , Oxidative Phosphorylation/drug effects , Primaquine/toxicity , Quinine/toxicity , Rats , Rats, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL