Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 343
Filter
1.
J Biochem Mol Toxicol ; 37(7): e23356, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37009961

ABSTRACT

Zidovudine (AZT) is the most commonly prescribed antiviral drug for the treatment of human immunodeficiency virus (HIV) infection. However, its chronic administration causes toxic side effects limiting its use. This study aimed to evaluate the toxicity of different concentrations of AZT and novel chalcogen derivatives (7A, 7D, 7G, 7K, 7M) on locomotion, mitochondrial dysfunction, acetylcholinesterase (AChE) activity, and production of reactive oxygen species (ROS) in adult Drosophila melanogaster. Our results show that AZT and its derivative 7K at a concentration of 10 µM impaired flies' locomotor behavior. Furthermore, AZT and the derivatives 7K, 7A, and 7M induced mitochondrial dysfunction observed by a decrease in oxygen flux through mitochondrial complexes I and II. Neither of the compounds tested affected AChE activity or ROS production in flies. According to these data, AZT derivatives presented the following decreasing order of toxicity: 7K > AZT > 7G > 7A > 7M > 7D. Based on the chemical structure, it is possible to infer that the presence of the seleno-phenyl group in 7A and 7G increases their toxicity compared to compounds 7D and 7M. In addition, compounds 7G, 7M, and 7K with three carbon atoms as spacer were more toxic than analogs containing one carbon atom (7A and 7D). Finally, the insertion of a p-methoxyl group enhances toxicity (7K). Based on these results, excepting 7K, all other chalcogen derivatives presented lower toxicity than AZT and are potential drug candidates.


Subject(s)
Anti-HIV Agents , Chalcogens , Animals , Humans , Zidovudine/toxicity , Drosophila melanogaster , Reactive Oxygen Species , Acetylcholinesterase , Anti-HIV Agents/toxicity
2.
Basic Clin Pharmacol Toxicol ; 133(1): 82-97, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37016497

ABSTRACT

INTRODUCTION: A combination of zidovudine (AZT), lamivudine (3TC) and lopinavir/ritonavir (LPV/r) is one of the most effective drugs for preventing mother-to-child transmission (PMTCT) of HIV. However, limited information is available regarding its systemic toxicity. This study aimed to investigate its potential toxicity. METHOD: An acute oral toxicity test was conducted to assess the potential acute toxicity of AZT + 3TC + LPV/r. Bacterial reverse mutation, mammalian erythrocyte micronucleus and mouse spermatogonia chromosomal aberration tests were conducted to assess its potential genotoxicity. A 28-day feeding test was conducted to assess the potential subacute toxicity. RESULTS: In mice, the LD50 of the AZT + 3TC + LPV/r mixture was greater than 2000 mg/kg body weight (BW). The rate of micronucleated polychromatic erythrocytes (PCEs) increased in a dose-dependent manner in mice (P < 0.01). After treatment with AZT + 3TC + LPV/r for 28 days, the BW gain of male and female rats in the high-dose group was lower than that in the control group (P < 0.05); the relative weights of the liver, kidney, spleen and brain increased (P < 0.05); and pathological abnormalities appeared in the thyroid and spleen of male and female rats in the high-dose group. The haemoglobin (HGB) and red blood cells (RBCs) count in male and female rats decreased, but the white blood cells (WBCs) and lymphocyte apoptosis rates in male and female rats in the high-dose group increased (P < 0.05). The total protein, albumin, cholesterol and blood glucose levels of male and female rats in the high-dose group were significantly decreased (P < 0.05). The alanine aminotransferase (ALT), alkaline phosphatase (ALP), lactate dehydrogenase (LDH), creatinine (Cr) and blood urea nitrogen (BUN) levels of male and female rats in the medium- and high-dose groups increased significantly (P < 0.05). CONCLUSION: The results suggest that AZT + 3TC + LPV/r may exhibit genotoxicity and subacute toxicity under experimental conditions.


Subject(s)
Anti-HIV Agents , HIV Infections , Female , Male , Animals , Mice , Rats , Lamivudine/toxicity , Zidovudine/toxicity , Zidovudine/therapeutic use , Lopinavir/toxicity , Ritonavir , Anti-HIV Agents/toxicity , Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , Infectious Disease Transmission, Vertical/prevention & control , Mammals
3.
Toxicol Appl Pharmacol ; 450: 116170, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35843342

ABSTRACT

Multidrug resistance (MDR) transporters present in placenta and fetal tissues reduce intracellular accumulation of their substrates. Consequently, induction of protein expression may further reduce toxic effects of specific xenobiotics. This work aimed to study whether sustained drug treatments in utero could modulate MDR transporters P-gp, BCRP, and MRP2 and thus impact their fetoprotective action. Pregnant Sprague-Dawley rats were daily treated by gavage with zidovudine (AZT, 60 mg/kg) or lamivudine (3TC, 30 mg/kg) from gestation day (GD) 11 to 20. On GD 21, DNA damage and MDR protein abundance were assessed by comet assay and western blotting, respectively. Moreover, a single IV dose of AZT or 3TC was administered on GD 21 and drug concentrations were measured in maternal blood and fetal liver by HPLC-UV. Chronic exposure to 3TC caused significantly higher DNA damage than AZT in fetal liver cells, whereas no differences were observed in maternal blood cells. Increased levels of BCRP protein were found in the placenta and fetal liver after AZT, but not 3TC, chronic in utero exposure. Contrarily, no modifications in the protein abundance of P-gp or MRP2 were found after sustained exposure to these drugs. The area under the curve of AZT in fetal liver was significantly lower in the AZT-pretreated rats than in the VEH or 3TC groups. Moreover, pre-administration of the BCRP inhibitor gefitinib (20 mg/kg, IP) increased AZT levels to the values observed in the VEH-treated group in this tissue. On the other hand, the disposition of 3TC in maternal blood or fetal liver was not modified after chronic treatment in either group. In conclusion, chronic exposure to AZT selectively induces BCRP expression in the placenta and fetal liver decreasing its own accumulation which may account for the lower DNA damage observed for AZT compared to 3TC in fetal liver cells.


Subject(s)
Anti-HIV Agents , Reverse Transcriptase Inhibitors , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Animals , Anti-HIV Agents/pharmacology , Drug Resistance, Multiple , Female , Fetus , Lamivudine/toxicity , Neoplasm Proteins , Placenta , Pregnancy , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Inhibitors/pharmacology , Zidovudine/toxicity
4.
Environ Toxicol Pharmacol ; 81: 103540, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33161113

ABSTRACT

BACKGROUND/AIMS: Zidovudine (3'-azido-2',3'-deoxythymidine; AZT) is a first-line drug for treatment of human immunodeficiency virus infection (HIV). However, its application is limited by cardiotoxicity due to cardiomyocyte injury. This study investigated whether Aloe-emodin (AE), an anthraquinone compound, protects against AZT-induced cardiomyocyte toxicity. METHODS: MTT, JC-1 assays and TUNEL were examined to verify the protective effect of AE against AZT-induced cardiomyocyte injury. Western blotting was performed to explore the anti-apoptotic effect of AE using anti-apoptotic proteins p90rsk, p-bad, and bcl-2 and pro-apoptotic proteins apaf-1, cleaved-caspase-3, and cytochrome c. RESULTS: We observed a protective effect of AE against cell viability decrease and TUNEL positive cells increase induced by AZT, which was counteracted by BI-D1870. Western blot analysis found that AE significantly inhibited cardiomyocyte apoptosis by activating p90rsk/p-bad/bcl-2 signaling pathway. Furthermore, BI-D1870 counteracted the anti-apoptotic effect of AE. CONCLUSIONS: Taken together, these results indicate that AE attenuated AZT-induced cardiomyocyte apoptosis by activating p90rsk.


Subject(s)
Anthraquinones/pharmacology , Anti-HIV Agents/toxicity , Cardiotonic Agents/pharmacology , Myocytes, Cardiac/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Zidovudine/toxicity , bcl-Associated Death Protein/metabolism , Animals , Animals, Newborn , Cell Survival/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/physiology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Rats, Sprague-Dawley , Signal Transduction/drug effects
5.
Acta Pharmacol Sin ; 41(2): 181-191, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31142800

ABSTRACT

Breast cancer resistance protein (BCRP) is one of ATP-binding cassette (ABC) transporters in brain microvessel endothelial cells that transport their substrates from brain to blood, thus limiting substrates to crossing into brain through blood-brain barrier. Our previous works show that bile duct ligation (BDL) impairs expression and function of brain BCRP in rats. Since zidovudine (AZT) is BCRP substrate, we investigated whether impaired expression and function of BCRP increased brain distribution and toxicity of AZT in BDL-D7 rats. After administration of AZT (10 mg/kg, i.v.), BDL markedly increased brain AZT concentrations, compared with sham-operated (SO) rats. The ratio of AZT brain-to-plasma area under concentration curve (AUC) in BDL rats was increased to 1.6-folds of SO rats. After treatment with AZT (100 mg/kg every day, i.v.) for 7 days, BDL significantly impaired cognitive functions compared with SO rats, evidenced by the significantly decreased percentage of alternation in Y-maze test and prolonged escaped latency in two-way passive avoidance trial. Furthermore, AZT treatment caused significant decrease in copies of mitochondrial DNA and mitochondrial membrane potential in hippocampus of BDL rats. Moreover, AZT treatment caused a significant decrease of cortex microtubule-associated protein 2 and hippocampus synaptophysin levels in BDL rats. AZT-induced CNS adverse alterations in BDL rats were not observed in SO rats treated with AZT. In conclusion, BDL decreases the function and expression of brain BCRP in rats, leading to increased brain distribution of AZT, which in turn enhances AZT CNS toxicity, leading to mitochondrial dysfunction, neuronal damage, and ultimately cognitive dysfunction.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Anti-HIV Agents/toxicity , Brain/drug effects , Zidovudine/toxicity , Animals , Anti-HIV Agents/pharmacokinetics , Area Under Curve , Bile Ducts/pathology , Blood-Brain Barrier/metabolism , Brain/pathology , Cell Line , Cognition/drug effects , Cognitive Dysfunction/chemically induced , Dogs , Humans , Madin Darby Canine Kidney Cells , Male , Rats , Rats, Sprague-Dawley , Tissue Distribution , Zidovudine/pharmacokinetics
6.
Drug Chem Toxicol ; 43(4): 429-434, 2020 Jul.
Article in English | MEDLINE | ID: mdl-30208744

ABSTRACT

Highly active antiretroviral therapy (HAART) regimens are based on the use of nucleoside reverse transcriptase inhibitors (NRTIs), which are the main drugs used by patients infected with the human immunodeficiency virus (HIV). The use of NRTIs combinations has afforded clear clinical benefits to patients undergoing HAART. However, the combination of two NRTIs may increase the risk of genomic instability in comparison with the drugs administered individually. We analyzed the ability of zidovudine (AZT) and lamivudine (3TC), and the combination AZT +3TC to induce complex genomic alterations using the cytokinesis-block micronucleus (CBMN) assay in Chinese hamster ovary (CHO)-K1 cells. The 24-h cell treatment with individual NRTIs showed that AZT increased micronucleus frequencies and nucleoplasmic bridges (NPBs). No significant differences were observed for any parameters investigated after exposure of CHO-K1 cells to 3TC. The combination AZT +3TC significantly increased micronucleus frequencies. Analysis of interaction between these drugs suggested that antagonism occurs in all AZT +3TC concentrations. These results highlight the importance to investigate the genotoxic profile of NRTIs to develop safer intervention strategies in antiretroviral treatment protocols.


Subject(s)
Anti-HIV Agents/toxicity , Antiretroviral Therapy, Highly Active/adverse effects , DNA Damage , Lamivudine/toxicity , Reverse Transcriptase Inhibitors/toxicity , Zidovudine/toxicity , Animals , CHO Cells , Cricetulus , Lamivudine/administration & dosage , Mutagenesis , Mutation , Zidovudine/administration & dosage
7.
Environ Mol Mutagen ; 60(5): 404-409, 2019 06.
Article in English | MEDLINE | ID: mdl-29206312

ABSTRACT

All nucleoside analogues for treating HIV infection, due to their capacity to integrate into and alter human DNA, are experimentally genotoxic to some extent. The long-term oncogenic risk after in utero exposure remains to be determined. Cancer incidence in uninfected children exposed to nucleos(t)ide reverse transcriptase inhibitors (NRTIs) was evaluated, by cross-checking against the National Cancer Registry, in the French perinatal study of children born to HIV+ mothers. Twenty-one cancers were identified in 15,163 children (median age: 9.9 years [interquartile range (IQR): 5.8-14.2]) exposed to at least one NRTI in utero between 1990 and 2014. Five of these children were exposed to zidovudine monotherapy, and 15 to various combinations, seven of which included didanosine. Overall, the total number of cases was not significantly different from that expected for the general population (SIR = 0.8[0.47-1.24]), but the number of cases after didanosine exposure was twice that expected (SIR = 2.5 [1.01-5.19]). Didanosine accounted for only 10% of prescriptions but was associated with one-third of cancers. In multivariate analysis, didanosine exposure was significantly associated with higher risk (HR = 3.0 [0.9-9.8]). This risk was specifically linked to first-trimester exposure (HR = 5.5 [2.1-14.4]). Three cases of pineoblastoma, a very rare cancer, were observed, whereas 0.03 were expected. Two were associated with didanosine exposure. Despite reassuring data overall, there is strong evidence to suggest that didanosine displays transplacental oncogenicity. These findings cannot be extrapolated to other NRTIs, but they highlight the need for comprehensive evaluations of the transplacental genotoxicity of this antiretroviral class. Environ. Mol. Mutagen., 60:404-409, 2019. © 2017 Wiley Periodicals, Inc.


Subject(s)
Anti-HIV Agents/toxicity , Maternal Exposure , Maternal-Fetal Exchange/physiology , Neoplasms/epidemiology , Nucleosides/toxicity , Prenatal Exposure Delayed Effects/pathology , Adolescent , Anti-HIV Agents/therapeutic use , Child , Child, Preschool , Didanosine/therapeutic use , Didanosine/toxicity , Female , HIV Infections/drug therapy , Humans , Incidence , Infant , Infant, Newborn , Male , Neoplasms/genetics , Nucleosides/therapeutic use , Pregnancy , Prospective Studies , Reverse Transcriptase Inhibitors/therapeutic use , Reverse Transcriptase Inhibitors/toxicity , Risk , Surveys and Questionnaires , Zidovudine/therapeutic use , Zidovudine/toxicity
8.
Hum Exp Toxicol ; 38(4): 446-454, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30545272

ABSTRACT

Abacavir (ABC), zidovudine (AZT), and lamivudine (3TC) are nucleoside analog reverse transcriptase inhibitors (NRTIs) widely used as combination-based antiretroviral therapy against human immunodeficiency virus. Despite effective viral suppression using NRTI combinations, genotoxic potential of NRTIs can be increased when administered in combination. This study investigated the toxic and genotoxic potential of ABC when administered alone or in combination with AZT and/or 3TC using the somatic mutation and recombination test in Drosophila melanogaster. This test simultaneously evaluated two events related to carcinogenic potential: mutation and somatic recombination. The results indicated that ABC was responsible for toxicity when administered alone or in combination with AZT and/or 3TC. In addition, all treatment combinations increased frequencies of mutation and somatic recombination. The combination of AZT/3TC showed the lowest genotoxic activity compared to all combinations with ABC. Therefore, our results indicated that ABC was responsible for a significant portion of genotoxic activity of these combinations. Somatic recombination was the main genetic event observed, ranging from 83.7% to 97.7%.


Subject(s)
Anti-HIV Agents/toxicity , Dideoxynucleosides/toxicity , Drosophila melanogaster/drug effects , Lamivudine/toxicity , Zidovudine/toxicity , Animals , DNA Damage , Drosophila melanogaster/genetics , Drug Synergism , Mutation , Recombination, Genetic
9.
Article in English | MEDLINE | ID: mdl-30373793

ABSTRACT

Nucleoside reverse transcriptase inhibitors (NRTI), such as zidovudine (AZT), are constituents of HIV-1 therapy and are used for the prevention of mother-to-child transmission. Prolonged thymidine analogue exposure has been associated with mitochondrial toxicities to heart, liver, and skeletal muscle. We hypothesized that the thymidine analogue AZT might interfere with autophagy in myocytes, a lysosomal degradation pathway implicated in the regulation of mitochondrial recycling, cell survival, and the pathogenesis of myodegenerative diseases. The impact of AZT and lamivudine (3TC) on C2C12 myocyte autophagy was studied using various methods based on LC3-green fluorescent protein overexpression or LC3 staining in combination with Western blotting, flow cytometry, and confocal and electron microscopy. Lysosomal and mitochondrial functions were studied using appropriate staining for lysosomal mass, acidity, cathepsin activity, as well as mitochondrial mass and membrane potential in combination with flow cytometry and confocal microscopy. AZT, but not 3TC, exerted a significant dose- and time-dependent inhibitory effect on late stages of autophagosome maturation, which was reversible upon mTOR inhibition. Inhibition of late autophagy at therapeutic drug concentrations led to dysfunctional mitochondrial accumulation with membrane hyperpolarization and increased reactive oxygen species (ROS) generation and, ultimately, compromised cell viability. These AZT effects could be readily replicated by pharmacological and genetic inhibition of myocyte autophagy and, most importantly, could be rescued by pharmacological stimulation of autophagolysosomal biogenesis. Our data suggest that the thymidine analogue AZT inhibits autophagy in myocytes, which in turn leads to the accumulation of dysfunctional mitochondria with increased ROS generation and compromised cell viability. This novel mechanism could contribute to our understanding of the long-term side effects of antiviral agents.


Subject(s)
Autophagy/drug effects , Lamivudine/toxicity , Mitochondria/pathology , Muscle Cells/pathology , Reverse Transcriptase Inhibitors/toxicity , Zidovudine/toxicity , Anti-HIV Agents/pharmacology , Cell Line , Cell Survival/drug effects , Humans , Infectious Disease Transmission, Vertical/prevention & control , Reactive Oxygen Species/metabolism
10.
Arch Toxicol ; 92(6): 2109-2118, 2018 06.
Article in English | MEDLINE | ID: mdl-29594326

ABSTRACT

The nucleoside reverse transcriptase inhibitor zidovudine (AZT), used in HIV infection treatment, induces mitochondrial DNA (mtDNA) depletion. A cause-effect relationship between mtDNA status alterations and autophagy has been reported. Both events are common in several liver diseases, including hepatocellular carcinoma. Here, we have studied autophagy activation in rat liver with mtDNA depletion induced by AZT administration in drinking water for 35 days. AZT at a concentration of 1 mg/ml, but not 0.5 mg/ml in the drinking water, decreased mtDNA levels in rat liver and extrahepatic tissues. In liver, mtDNA-encoded cytochrome c oxidase 1 protein levels were decreased. Although serum biomarkers of liver and kidney toxicity remained unaltered, ß-hydroxybutyrate levels were increased in liver of AZT-treated rats. Moreover, autophagy was dysregulated at two levels: (i) decreased induction signalling of this process as indicated by increases in autophagy inhibitors activity (AKT/mTOR), and absence of changes (Beclin-1, Atg5, Atg7) or decreases (AMPK/ULK1) in the expression/activity of pro-autophagy proteins; and (ii) reduced autophagosome degradation as indicated by decreases in the lysosome abundance (LAMP2 marker) and the transcription factor TFEB controlling lysosome biogenesis. This resulted in increased autophagosome abundance (LC3-II marker) and accumulation of the protein selectively degraded by autophagy p62, and the transcription factor Nrf2 in liver of AZT-treated rats. Nrf2 was activated as indicated by the up-regulation of antioxidant target genes Nqo1 and Hmox-1. In conclusion, rat liver with AZT-induced mtDNA depletion presented dysregulations in autophagosome formation and degradation balance, which results in accumulation of these structures in parenchymal liver cells, favouring hepatocarcinogenesis.


Subject(s)
Autophagosomes/drug effects , Autophagy/drug effects , DNA, Mitochondrial/drug effects , Liver/drug effects , Reverse Transcriptase Inhibitors/toxicity , Zidovudine/toxicity , Animals , Autophagosomes/pathology , Cell Line, Tumor , Liver/pathology , Male , Rats, Wistar
11.
J Hazard Mater ; 349: 195-204, 2018 05 05.
Article in English | MEDLINE | ID: mdl-29427970

ABSTRACT

The concentration of antiretroviral drugs in wastewater treatment plants (WWTP) effluents and surface waters of many countries has increased significantly due to their widespread use for HIV treatment. In this study, the removal of stavudine and zidovudine under UV254 photolysis or UV254/H2O2 was investigated in a microcapillary film (MCF) photoreactor, using minimal water samples quantities. The UV254 quantum yield of zidovudine, (2.357 ±â€¯0.0589)·10-2 mol ein-1 (pH 4.0-8.0), was 28-fold higher that the yield of stavudine (8.34 ±â€¯0.334)·10-4 mol ein-1 (pH 6.0-8.0). The second-order rate constant kOH,iof reaction of hydroxyl radical with the antiretrovirals (UV254/H2O2 process) were determined by kinetics modeling: (9.98 ±â€¯0.68)·108 M-1 s-1 (pH 4.0-8.0) for zidovudine and (2.03 ±â€¯0.18)·109 M-1 s-1 (pH 6.0-8.0) for stavudine. A battery of ecotoxicological tests (i.e. inhibition growth, bioluminescence, mutagenic and genotoxic activity) using bacteria (Aliivibrio fischeri, Salmonella typhimurium), crustacean (Daphnia magna) and algae (Raphidocelis subcapitata) revealed a marked influence of the UV dose on the ecotoxicological activity. The UV254/H2O2 treatment process reduced the ecotoxicological risk associated to direct photolysis of the antiretrovirals aqueous solutions, but required significantly higher UV254 doses (≥2000 mJ cm-2) in comparison to common water UV disinfection processes.


Subject(s)
Anti-Retroviral Agents , Hydrogen Peroxide , Stavudine , Ultraviolet Rays , Water Pollutants, Chemical , Zidovudine , Aliivibrio fischeri/drug effects , Aliivibrio fischeri/growth & development , Animals , Anti-Retroviral Agents/chemistry , Anti-Retroviral Agents/toxicity , Daphnia/drug effects , Daphnia/physiology , Ecotoxicology , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/radiation effects , Kinetics , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Stavudine/chemistry , Stavudine/toxicity , Water Pollutants, Chemical/chemistry , Water Pollutants, Chemical/toxicity , Zidovudine/chemistry , Zidovudine/toxicity
12.
Toxicol Sci ; 160(1): 30-46, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29036705

ABSTRACT

Considering a novel series of zidovudine (AZT) derivatives encompassing selenoaryl moieties promising candidates as therapeutics, we examined the toxicities elicited by AZT and derivatives 5'-(4-Chlorophenylseleno)zidovudine (SZ1); 5'-(Phenylseleno)zidovudine (SZ2); and 5'-(4-Methylphenylseleno)zidovudine (SZ3) in healthy cells and in mice. Resting and stimulated cultured human peripheral blood mononuclear cells (PBMCs) were treated with the compounds at concentrations ranging from 10 to 200 µM for 24 and/or 72 h. Adult mice received a single injection of compounds (100 µmol/kg, s.c.) and 72 h after administration, hepatic/renal biomarkers were analyzed. Resting and stimulated PBMCs exposed to SZ1 displayed loss of viability, increased reactive species production, disruption in cell cycle, apoptosis and increased transcript levels and production of pro-inflammatory cytokines. In a mild way, most of these effects were also induced by SZ2. AZT and SZ3 did not cause significant toxicity towards resting PBMCs. Differently, both compounds elicited apoptosis and S phase arrest in stimulated cells. AZT and derivatives administration did not change the body weight and plasma biochemical markers in mice. However, the absolute weight and organ-to-body weight ratio of liver, kidneys and spleen were altered in AZT, SZ1-, and SZ2-treated mice. Our results highlighted the involvement of derivatives SZ1 and SZ2 in redox and immunological dyshomeostasis leading to activation of apoptotic signaling pathways in healthy cells under different division phases. On the other hand, the derivative SZ3 emerged as a promising candidate for further viral infection/antitumor studies as a new effective therapy with low toxicity for immune cells and after acute in vivo treatment.


Subject(s)
Antineoplastic Agents/toxicity , Chalcogens/toxicity , Leukocytes, Mononuclear/drug effects , Zidovudine/toxicity , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Behavior, Animal/drug effects , Cell Survival/drug effects , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation , Humans , Inflammation Mediators/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Organ Size/drug effects , Reactive Oxygen Species/metabolism , Risk Assessment , S Phase Cell Cycle Checkpoints/drug effects , Zidovudine/analogs & derivatives
13.
Chem Phys Lipids ; 207(Pt A): 24-37, 2017 10.
Article in English | MEDLINE | ID: mdl-28698149

ABSTRACT

The major challenges to clinical application of zidovudine are its moderate aqueous solubility and relative short half-life and serious side effects due to frequent administrations. We investigated the preparation of zidovudine-loaded nanoparticles based on lipids which were further modified with the polymer gelatin. Formulation and stability of the modified nanoparticles were analysed from the physico-chemical characterizations. The interactions of nanoparticles with blood components were tested by haemolysis and aggregation studies. The drug content and entrapment efficiencies were assessed by UV analysis. The effect of nanoparticles on protein adsorption was assessed by native polyacrylamide gel electrophoresis (PAGE). In vitro release studies showed a sustained release profile of zidovudine. In vitro cytotoxicity and cellular uptake of the zidovudine-loaded nanoparticles were performed in MCF-7 and neuro 2a brain cells. The enhanced cellular internalization of drug loaded modified nanoparticles in both the cell lines were revealed by fluorescence microscopy. Hence the present study focuses on the feasibility of zidovudine-loaded polymer modified lipid nanoparticles as carriers for safe and efficient HIV/AIDS therapy.


Subject(s)
Antiviral Agents/chemistry , Drug Carriers/chemistry , Gelatin/chemistry , Lipids/chemistry , Nanoparticles/chemistry , Adsorption , Antiviral Agents/metabolism , Antiviral Agents/toxicity , Blood Proteins/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers/toxicity , Drug Compounding , Drug Liberation , Drug Stability , Humans , MCF-7 Cells , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Particle Size , Spectroscopy, Fourier Transform Infrared , Zidovudine/chemistry , Zidovudine/metabolism , Zidovudine/toxicity
14.
Redox Biol ; 13: 407-417, 2017 10.
Article in English | MEDLINE | ID: mdl-28683400

ABSTRACT

Highly active anti-retrovirus therapy (HAART) has been used to block the progression and symptoms of human immunodeficiency virus infection. Although it decreases morbidity and mortality, clinical use of HAART has also been linked to various adverse effects such as severe cardiomyopathy resulting from compromised mitochondrial functioning. However, the mechanistic basis for these effects remains unclear. Here, we demonstrate that a key component of HAART, 3ꞌ-azido-3ꞌ-deoxythymidine (AZT), particularly, its active metabolite AZT-triphosphate (AZT-TP), caused mitochondrial dysfunction, leading to induction of cell death in H9c2 cells derived from rat embryonic myoblasts, which serve as a model for cardiomyopathy. Specifically, treatment with 100µM AZT for 48h disrupted the mitochondrial tubular network via accumulation of AZT-TP. The mRNA expression of dynamin-related protein (Drp)1 and the Drp1 receptor mitochondrial fission factor (Mff) was upregulated whereas that of optic atrophy 1 (Opa1) was downregulated following AZT treatment. Increased mitochondrial translocation of Drp1, Mff upregulation, and decreased functional Opa1 expression induced by AZT impaired the balance of mitochondrial fission vs. fusion. These data demonstrate that AZT-TP causes cell death by altering mitochondrial dynamics.


Subject(s)
Anti-HIV Agents/toxicity , Mitochondria/drug effects , Mitochondrial Dynamics , Zidovudine/toxicity , Animals , Anti-HIV Agents/adverse effects , Cardiotoxicity/etiology , Cell Line , Dynamins/genetics , Dynamins/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Mitochondria/metabolism , Rats , Zidovudine/adverse effects
15.
Chem Biol Interact ; 273: 142-153, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28619387

ABSTRACT

HIV/AIDS patients have suppressed immune system, making them vulnerable to many opportunistic infections including tuberculosis (TB). The patients who are co-infected with TB undergo combined regimens with anti-retroviral drugs such as zidovudine (AZT) and anti-tubercular drug such as isoniazid (INH) for therapy leading to hepatotoxicty. Silibinin (SBN), extracted from Silybum marianum commonly called as "Milk thistle" is used against several drugs-induced hepatotoxicity. The present study evaluates the ameliorative effect of SBN against AZT alone, INH alone, and INH + AZT-induced toxic insults to liver of rats. Wistar albino rats (n = 6/groups) were given INH and AZT (25 and 50 mg mg/kg b.w.) respectively either alone or in combination for a sub-chronic period of 45 days orally. Another group of rats received SBN (100 mg/kg b.w.) along with INH and AZT. The group that received propylene glycol served as control. AZT alone, INH alone and INH + AZT treatments showed parenchymal cell injury and cholestasis by highly significant increase in the activities of marker enzymes (aspartate and alanine transaminase, alkaline phosphatase, argino succinic acid lyase), bilirubin and protein. The presence of hyperlipidaemia was observed by analyzing lipid profiles in serum/liver/adipose tissue, gene expression (RT-PCR) of Phase-I and II metabolizing enzymes and western blot. Transmission electron microscopy study also revealed large vacuoles with membraneous debri, pleomorphic mitochondria, disruption of endoplasmic reticulum, presence of lipid droplets, breakage in cellular and nuclear membrane. SBN simultaneous treatment showed ameliorative effect against INH + AZT-induced hepatotoxicity and hyperlipidemia in rats.


Subject(s)
Hyperlipidemias/drug therapy , Isoniazid/toxicity , Liver/drug effects , Metabolic Detoxication, Phase II , Metabolic Detoxication, Phase I , Silymarin/pharmacology , Zidovudine/toxicity , Adipose Tissue/drug effects , Adipose Tissue/pathology , Animals , Dose-Response Relationship, Drug , Female , Hyperlipidemias/pathology , Isoniazid/metabolism , Lipids/analysis , Liver/pathology , Male , Rats , Rats, Wistar , Silybin , Silymarin/administration & dosage , Silymarin/metabolism , Silymarin/therapeutic use , Structure-Activity Relationship , Zidovudine/metabolism
16.
Hum Exp Toxicol ; 36(9): 910-918, 2017 Sep.
Article in English | MEDLINE | ID: mdl-27777322

ABSTRACT

Acquired immunodeficiency syndrome (AIDS) is a worldwide disease characterized by impairments of immune function. AIDS can be associated with oxidative stress (OS) that can be linked to selenium (Se) deficiency. Se is fundamental for the synthesis of selenoproteins, such as glutathione peroxidase and thioredoxin reductase. These enzymes catalyze the decomposition of reactive oxygen species and contribute to maintain equilibrium in cell redox status. Literature data indicate that organoselenium compounds, such as ebselen and diphenyl diselenide, have antioxidant properties in vitro and in vivo models associated with OS. Nevertheless, selenocompounds can also react and oxidize thiols groups, inducing toxicity in mammals. Here, we tested the potential cytotoxic and genotoxic properties of six analogs of the prototypal anti-HIV drug azidothymidine (AZT) containing Se (5'-Se-(phenyl)zidovudine; 5'-Se-(1,3,5-trimethylphenyl)zidovudine; 5'-Se-(1-naphtyl)zidovudine; 5'-Se-(4-chlorophenyl)zidovudine) (C4); 5'-Se-(4-methylphenyl)zidovudine (C5); and 5'-(4-methylbenzoselenoate)zidovudine). C5 increased the rate of dithiothreitol oxidation (thiol oxidase activity) and C2-C4 and C6 (at 100 µM) increased DNA damage index (DI) in human leukocytes. Moreover, C5 (200 µM) decreased human leukocyte viability to about 50%. Taken together, these results indicated the low in vitro toxicity in human leukocytes of some Se-containing analogs of AZT.


Subject(s)
Anti-HIV Agents/toxicity , Leukocytes/drug effects , Organoselenium Compounds/toxicity , Zidovudine/analogs & derivatives , Zidovudine/toxicity , Cell Survival/drug effects , Cells, Cultured , Comet Assay , DNA Damage , Humans
17.
Article in English | MEDLINE | ID: mdl-27931817

ABSTRACT

In vivo phosphatidylinositol glycan, class A (Pig-a) gene mutation assay using peripheral blood is known to be a novel and useful tool to evaluate the mutagenicity of compounds. Recently, the rat PIGRET assay which is an improved method for measuring Pig-a mutant cells in reticulocytes with magnetic enrichment of CD71 positive cells has been developed. Several reports showed that the PIGRET assay could detect the increase of Pig-a mutant frequency earlier than the Pig-a assay in total red blood cells (RBC Pig-a assay). Therefore, as part of a collaborative study by the Mammalian Mutagenicity Study (MMS) Group of the Japanese Environmental Mutagen Society, the usefulness of the PIGRET assay in comparison to the RBC Pig-a assay has been assessed for 24 compounds with various mechanisms of action. In the present study, we performed the PIGRET assay and RBC Pig-a assay with a nucleoside analogue, azidothymidine (AZT), and compared the results in these assays. We administered a single dose of AZT to rats by oral gavage up to 2000mg/kg and examined Pig-a mutant frequencies at days 7, 14 and 28 by PIGRET and RBC Pig-a assays. No significant increases in mutant frequency were observed after administration of AZT in both the RBC Pig-a and PIGRET assays and comparable to the previous results of the International Workshop on Genotoxicity Testing (IWGT) workgroup. AZT has been thought to induce not only DNA chain termination as a pharmacological effect but also a large deletion on the genome DNA. The Pig-a assays may be less sensitive to compounds such as AZT which induce large deletions on the genome DNA.


Subject(s)
Anti-HIV Agents/toxicity , Mutagenicity Tests/methods , Mutagens/toxicity , Reticulocytes/drug effects , Zidovudine/toxicity , Administration, Oral , Animals , Anti-HIV Agents/administration & dosage , Membrane Proteins/genetics , Mutagens/administration & dosage , Rats , Zidovudine/administration & dosage
18.
Environ Toxicol Pharmacol ; 46: 217-226, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27497728

ABSTRACT

HIV/AIDS patients are more prone for opportunistic TB infections and they are administered the combined regimen of anti-retroviral drug zidovudine (AZT) and isoniazid (INH) for therapy. However, AZT+INH treatment has been documented to induce injury and remedial measures to prevent this adversity are not clearly defined. Silibinin (SBN) is a natural hepatoprotective principle isolated from medicinal plant Silybum marianum and is currently used for therapy of various liver diseases. This study investigate the hepatotoxic potentials of AZT alone, INH alone and AZT+INH treatments and the mitigating potentials of SBN against these drugs induced toxic insults of liver in rats. Separate groups of rats (n=6 in each group) were administered AZT alone (50mg/kg b.w.), INH alone (25mg/kg, b.w.), AZT+INH (50mg/kg, b.w. and 25mg/kg, b.w.), SBN alone (100mg/kg, b.w.) and SBN+AZT+INH daily for sub-chronic period of 45days orally. The control rats received saline/propylene glycol. INH alone and AZT+INH-induced parenchymal cell injury and cholestasis of liver was evidenced by highly significant increase in the activities of marker enzymes (aspartate and alanine transaminase, alkaline phosphatase, argino succinic acid lyase), bilirubin, protein, oxidative stress parameters (lipid peroxidation, superoxide dismutase, catalase, reduced glutathione, vitamins C and E) and membrane bound ATPases were evaluated in serum/liver tissue homogenates. Histopathological studies show ballooning degradation, inflammatory lesions, lipid deposition and hydropic changes in the liver tissue. All the above biochemical and pathological changes induced by AZT+INH treatments were mitigated in rats receiving SBN simultaneously with these hepatotoxins, indicating its hepatoprotective and antioxidant potentials against AZT+INH-induced hepatotoxicity. The moderate hepatoprotective and oxidant potentials of SBN could be due to its low bioavailability and this deficiency could be prevented by supplementation of phosphatidylcholines and studies are warranted on these lines to improve the therapeutic efficiency of SBN.


Subject(s)
Antioxidants/therapeutic use , Chemical and Drug Induced Liver Injury/prevention & control , Isoniazid/toxicity , Oxidative Stress/drug effects , Silymarin/therapeutic use , Zidovudine/toxicity , Animals , Antioxidants/administration & dosage , Biomarkers/blood , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Drug Synergism , Female , Isoniazid/administration & dosage , Liver Function Tests , Male , Rats, Wistar , Silybin , Silymarin/administration & dosage , Zidovudine/administration & dosage
19.
Environ Mol Mutagen ; 57(7): 526-34, 2016 08.
Article in English | MEDLINE | ID: mdl-27452341

ABSTRACT

Antiretroviral (ARV) drug therapy, given during pregnancy for prevention of mother-to-child transmission of human immunodeficiency virus 1 (HIV-1), induces fetal mitochondrial dysfunction in some children. However, the persistence/reversibility of that dysfunction is unclear. Here we have followed Erythrocebus patas (patas) monkey offspring for up to 3 years of age (similar in development to a 15-year old human) after exposure of the dams to human-equivalent in utero ARV exposure protocols. Pregnant patas dams (3-5/exposure group) were given ARV drug combinations that included zidovudine (AZT)/lamivudine (3TC)/abacavir (ABC), or AZT/3TC/nevirapine (NVP), for the last 10 weeks (50%) of gestation. Infants kept for 1 and 3 years also received drug for the first 6 weeks of life. In offpsring at birth, 1 and 3 years of age mitochondrial morphology, examined by electron microscopy (EM), was compromised compared to the unexposed controls. Mitochondrial DNA (mtDNA), measured by hybrid capture chemiluminescence assay (HCCA) was depleted in hearts of patas exposed to AZT/3TC/NVP at all ages (P < 0.05), but not in those exposed to AZT/3TC/ABC at any age. Compared to unexposed controls, mitochondrial reserve capacity oxygen consumption rate (OCR by Seahorse) in cultured bone marrow mesenchymal fibroblasts from 3-year-old patas offspring was ∼50% reduced in AZT/3TC/ABC-exposed patas (P < 0.01), but not in AZT/3TC/NVP-exposed patas. Overall the data show that 3-year-old patas sustain persistent mitochondrial dysfunction as a result of perinatal ARV drug exposure. Environ. Mol. Mutagen. 57:526-534, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Anti-HIV Agents/toxicity , DNA, Mitochondrial/analysis , Mitochondria/drug effects , Prenatal Exposure Delayed Effects/chemically induced , Animals , Anti-HIV Agents/administration & dosage , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Brain/drug effects , Brain/growth & development , Brain/pathology , DNA, Mitochondrial/genetics , Dideoxynucleosides/administration & dosage , Dideoxynucleosides/toxicity , Drug Therapy, Combination , Erythrocebus patas , Female , Gestational Age , Heart/drug effects , Heart/growth & development , Lamivudine/administration & dosage , Lamivudine/toxicity , Mitochondria/genetics , Mitochondria/ultrastructure , Mitochondria, Heart/drug effects , Mitochondria, Heart/genetics , Mitochondria, Heart/ultrastructure , Oxygen Consumption/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/pathology , Zidovudine/administration & dosage , Zidovudine/toxicity
20.
Toxicol In Vitro ; 35: 66-76, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27238871

ABSTRACT

Zidovudine (AZT) remains the mainstay of antiretroviral therapy against HIV in resource-poor countries; however, its use is frequently associated with hepatotoxicity. Not all HIV patients on AZT develop hepatotoxicity, and the determining factors are unclear. Alcohol consumption and cigarette smoking are known risk factors for HIV hepatotoxicity, and both are significant sources of acrolein, a highly reactive and toxic aldehyde. This study examines the potential hepatotoxic interactions between acrolein and AZT. Our data demonstrate that acrolein markedly enhanced AZT-induced transcriptionally permissive histone modifications (H3K9Ac and H3K9Me3) allowing the recruitment of transcription factor NF-kB and RNA polymerase II at the FasL gene promoter, resulting in FasL upregulation and apoptosis in hepatocytes. Notably, the acrolein scavenger, hydralazine prevented these promoter-associated epigenetic changes and inhibited FasL upregulation and apoptosis induced by the combination of AZT and acrolein, as well as AZT alone. Our data strongly suggest that acrolein enhancement of promoter histone modifications and FasL upregulation are major pathogenic mechanisms driving AZT-induced hepatotoxicity. Moreover, these data also indicate the therapeutic potential of hydralazine in mitigating AZT hepatotoxicity.


Subject(s)
Acrolein/toxicity , Anti-HIV Agents/toxicity , Epigenesis, Genetic/drug effects , Fas Ligand Protein/genetics , Hepatocytes/drug effects , Zidovudine/toxicity , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , DNA Fragmentation , Hep G2 Cells , Hepatocytes/metabolism , Histones/genetics , Humans , Hydralazine/pharmacology , RNA Polymerase II/genetics , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...