Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Biochem Biophys Res Commun ; 584: 46-52, 2021 12 20.
Article in English | MEDLINE | ID: mdl-34768081

ABSTRACT

Efavirenz (EFV) is a non-nucleoside reverse transcriptase inhibitor (NNRTI), which is widely used for anti-HIV-1. Evidences revealed that several central nervous system side effects could be observed in mice and patients with administration of EFV. However, the detailed mechanisms are still unknown. In this study, we investigated the effects of long-term EFV treatment on cognitive functions and the potential underlying mechanisms in mice. We maintained C57BL/6 mice aged 2 months with treatment containing 40 or 80 mg/kg/day EFV for 5 months, while control group treated with saline. The cognitive functions were evaluated by novel object recognition test, Barnes maze test and Morris water maze. The results showed significant short-term memory impairment in 40 and 80 mg/kg groups, and notable spatial learning and memory impairments in 80 mg/kg group, without any spontaneous activity alteration. Moreover, EFV induced impairments in dendritic integrity and synaptic plasticity in hippocampus. Furthermore, Significant increases were observed in the expression levels of pro-IL-1ß, a similar tendency of TNF-α and phosphorylation of p65 of the 80 mg/kg group compared with control group. These results imply that long-term EFV treatment causes synaptic dysfunction resulting in cognitive deficits, which might be induced by the enhanced pro-inflammatory cytokines IL-1ß and TNF-α via activating NF-κB pathway.


Subject(s)
Alkynes/toxicity , Benzoxazines/toxicity , Cognition/drug effects , Cognitive Dysfunction/physiopathology , Cyclopropanes/toxicity , Memory Disorders/physiopathology , Neuroinflammatory Diseases/physiopathology , Animals , Cognition/physiology , Cognitive Dysfunction/chemically induced , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/chemically induced , Mice, Inbred C57BL , Neuroinflammatory Diseases/chemically induced , Reverse Transcriptase Inhibitors/toxicity , Spatial Learning/drug effects , Spatial Learning/physiology , Synapsins/metabolism , Synaptophysin/metabolism , Synaptotagmin I/metabolism , Time Factors
2.
J Neurovirol ; 26(5): 642-651, 2020 10.
Article in English | MEDLINE | ID: mdl-32737860

ABSTRACT

Highly active antiretroviral treatment has led to unprecedented efficacy and tolerability in people living with HIV. This effect was also observed in the central nervous system with the nowadays uncommon observation of dementias; yet in more recent works milder forms are still reported in 20-30% of optimally treated individuals. The idea of a subclinical neuronal toxicity induced by antiretrovirals has been proposed and was somehow supported by the late-emerging effects associated with efavirenz use. In this manuscript we are reviewing all the potential mechanisms by which antiretroviral drugs have been associated with in vitro, ex vivo, or in vivo toxicity to cells pertaining to the central nervous system (neurons, astrocytes, oligodendrocytes, and endothelial cells). These include direct or indirect effects and pathological pathways such as amyloid deposition, damage to small cerebral vessels, and impairment in neurotransmission. The aim of this review is therefore to provide a detailed description of the available literature in order to guide further clinical research for improving patients' neurocognition and quality of life.


Subject(s)
Alkynes/toxicity , Anti-HIV Agents/toxicity , Benzoxazines/toxicity , Central Nervous System/drug effects , Cognitive Dysfunction/chemically induced , Cyclopropanes/toxicity , HIV Infections/drug therapy , Neurons/drug effects , Antiretroviral Therapy, Highly Active/methods , Astrocytes/drug effects , Astrocytes/pathology , Astrocytes/virology , Atazanavir Sulfate/toxicity , Central Nervous System/pathology , Central Nervous System/virology , Cognitive Dysfunction/pathology , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/virology , Dideoxynucleosides/toxicity , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelial Cells/virology , HIV Infections/pathology , HIV Infections/virology , Humans , Neurons/pathology , Neurons/virology , Nevirapine/toxicity , Nitriles/toxicity , Oligodendroglia/drug effects , Oligodendroglia/pathology , Oligodendroglia/virology , Pyrimidines/toxicity
3.
Cephalalgia ; 40(1): 68-78, 2020 01.
Article in English | MEDLINE | ID: mdl-31311288

ABSTRACT

AIM: Evaluation of cannabinoid receptor agonists in a preclinical model of medication overuse headache. METHODS: Female Sprague Dawley rats received graded intraperitoneal doses of WIN55,212-2 or Δ-9-tetrahydrocannabinol (Δ-9-THC). Antinociception (tail-flick test), catalepsy and hypomotility (open field test) and impairment of motor function (rotarod test) were assessed to establish effective dosing. Rats were then treated twice daily with equianalgesic doses of WIN55,212-2 or Δ-9-THC, or vehicle, for 7 days and cutaneous tactile sensory thresholds were evaluated during and three weeks following drug discontinuation. Rats then received a one-hour period of bright light stress (BLS) on two consecutive days and tactile sensory thresholds were re-assessed. RESULTS: WIN55,212-2 and Δ-9-THC produced antinociception as well as hypomotility, catalepsy and motor impairment. Repeated administration of WIN55,212-2 and Δ-9-THC induced generalized periorbital and hindpaw allodynia that resolved within 3 weeks after discontinuation of drug. Two episodes of BLS produced delayed and long-lasting periorbital and hindpaw allodynia selectively in rats previously treated with WIN55,212-2, and Δ-9-THC. INTERPRETATION: Cannabinoid receptor agonists including Δ-9-THC produce a state of latent sensitization characterized by increased sensitivity to stress, a presumed migraine trigger. Overuse of cannabinoids including cannabis may increase the risk of medication overuse headache in vulnerable individuals.


Subject(s)
Benzoxazines/toxicity , Cannabinoid Receptor Agonists/toxicity , Disease Models, Animal , Dronabinol/toxicity , Headache Disorders, Secondary/chemically induced , Morpholines/toxicity , Naphthalenes/toxicity , Pain Measurement/drug effects , Animals , Cannabinoids/toxicity , Dose-Response Relationship, Drug , Female , Headache Disorders, Secondary/psychology , Pain Measurement/methods , Rats , Rats, Sprague-Dawley
4.
Arch Toxicol ; 94(12): 4131-4141, 2020 12.
Article in English | MEDLINE | ID: mdl-32833042

ABSTRACT

In animals, research in the past two decades has demonstrated the strong involvement of the endocannabinoid system (ECS) in numerous steps of the reproductive process, including ovarian physiology. Reproductive lifespan is closely related to the number of nongrowing ovarian follicles, called ovarian reserve (OR), which is definitively established during foetal life. Thus, OR damage may lead to poor reproductive outcomes and a shortened reproductive lifespan. We investigated whether prenatal ECS modulation had an effect on the OR at different ages in the rat offspring. Four groups of gestating female rats (F0) were exposed to the CB1-/CB2-receptor agonist WIN55212 (0.5 mg/kg), the CB1R inverse agonist SR141716 (3 mg/kg) or Δ9THC (5 mg/kg) and were compared to negative control groups. OR was histologically assessed at different postnatal timepoints (F1 individuals): postnatal day (PND) 6, PND40 and PND90. At PND6, prenatal exposure had no effect on OR. In the young adult group (PND90) exposed during gestation to WIN55212, we observed a CB1R-mediated delayed OR decrease, which was reversed by prenatal CB1R blockade by SR141716. Conversely, after prenatal SR141716 exposure, we observed higher OR counts at PND90. RT-PCR experiments also showed that prenatal ECS modulation perturbed the mRNA levels of ECS enzymes and OR regulation genes. Our findings support the role of the ECS in OR regulation during the foetal life of rats and highlight the need for further studies to elucidate its precise role in OR physiology.


Subject(s)
Cannabinoid Receptor Agonists/toxicity , Dronabinol/toxicity , Ovarian Reserve/drug effects , Ovary/drug effects , Prenatal Exposure Delayed Effects , Receptor, Cannabinoid, CB1/agonists , Animals , Benzoxazines/toxicity , Cannabinoid Receptor Antagonists/pharmacology , Drug Inverse Agonism , Endocannabinoids/genetics , Endocannabinoids/metabolism , Female , Gene Expression Regulation , Gestational Age , Morpholines/toxicity , Naphthalenes/toxicity , Ovarian Reserve/genetics , Ovary/metabolism , Ovary/physiopathology , Pregnancy , Rats, Wistar , Receptor, Cannabinoid, CB1/metabolism , Rimonabant/pharmacology
5.
Biomed Chromatogr ; 34(8): e4860, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32311767

ABSTRACT

Cetilistat (CET) is a pancreatic lipase inhibitor approved for management of obesity after the serious adverse effects exhibited by its analogue orlistat. Exhaustive literature review reveals lack of comprehensive reports on its biotransformation. With a view to study the same, the present study reports the identification and characterization of metabolites of CET in rats using UPLC-MS/MS. As the small intestine is the site of action for CET, it is important that the role of microbial flora in the metabolism of CET be explored. To achieve this, the metabolic profile of CET was compared between normal and pseudo-germ-free rats. The study involved the administration of a drug suspension to male Sprague-Dawley pseudo-germ-free and normal untreated rats followed by collection of urine, feces, and blood at specific intervals. Sample preparation was performed using liquid-liquid extraction and concentration of samples followed by analysis using LC-MS/MS. Finally, an in silico study was performed on the drug and metabolites to predict their toxicological properties using ADMET PredictorTM software. Four metabolites of CET were observed in in vivo matrices. As expected, significant changes were observed both qualitatively and quantitatively, implying that formation of metabolites was both CYP enzymes and gut microflora mediated.


Subject(s)
Benzoxazines , Chromatography, High Pressure Liquid/methods , Tandem Mass Spectrometry/methods , Animals , Benzoxazines/blood , Benzoxazines/chemistry , Benzoxazines/pharmacokinetics , Benzoxazines/toxicity , Germ-Free Life , Male , Rats , Rats, Sprague-Dawley
6.
New Dir Child Adolesc Dev ; 2020(171): 107-133, 2020 May.
Article in English | MEDLINE | ID: mdl-32657046

ABSTRACT

Efavirenz (EFV) is a well-known, effective anti-retroviral drug long used in first-line treatment for children and adults with HIV and HIV/AIDS. Due to its narrow window of effective concentrations, between 1 and 4 µg/mL, and neurological side effects at supratherapeutic levels, several investigations into the pharmacokinetics of the drug and its genetic underpinnings have been carried out, primarily with adult samples. A number of studies, however, have examined the genetic influences on the metabolism of EFV in children. Their primary goal has been to shed light on issues of appropriate pediatric dosing, as well as the manifestation of neurotoxic effects of EFV in some children. Although EFV is currently being phased out of use for the treatment of both adults and children, we share this line of research to highlight an important aspect of medical treatment that is relevant to understanding the development of children diagnosed with HIV.


Subject(s)
Alkynes , Anti-HIV Agents , Benzoxazines , Child Development/drug effects , Cyclopropanes , Cytochrome P-450 CYP2B6/genetics , HIV Infections/drug therapy , Pharmacogenetics , Reverse Transcriptase Inhibitors , Alkynes/administration & dosage , Alkynes/metabolism , Alkynes/toxicity , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/metabolism , Anti-HIV Agents/toxicity , Benzoxazines/administration & dosage , Benzoxazines/metabolism , Benzoxazines/toxicity , Child , Child, Preschool , Cyclopropanes/administration & dosage , Cyclopropanes/metabolism , Cyclopropanes/toxicity , Humans , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/metabolism , Reverse Transcriptase Inhibitors/toxicity
7.
J Neurovirol ; 25(1): 85-90, 2019 02.
Article in English | MEDLINE | ID: mdl-30478800

ABSTRACT

The mechanisms underlying central nervous system (CNS) toxicities in antiretroviral-treated persons living with HIV (PLWH) remain elusive. We investigated the associations between markers of tryptophan metabolism and measurements of CNS toxicity in PLWH. In a prospective study, virologically suppressed PLWH receiving efavirenz-containing antiretroviral regimens with ongoing CNS toxicity were switched to dolutegravir-containing regimens and followed up for 12 weeks. Plasma tryptophan and kynurenine concentrations and the kynurenine/tryptophan ratio were calculated. Ten CNS toxicities were graded according to the ACTG adverse events scale. Scores ranged from 0 (none) to 3 (severe) and were summed, giving a total from 0 to 30. Paired-samples t tests and linear mixed model analyses were conducted to assess changes in, and relationships between, laboratory and clinical parameters. Mean kynurenine plasma concentration increased from baseline to week 12 (2.15 to 2.50 µmol/L, p = 0.041). No significant changes were observed for tryptophan (54.74 to 56.42 µmol/L, p = 1.000) or kynurenine/tryptophan ratio (40.37 to 41.08 µmol/L, p = 0.276). Mean CNS toxicity score decreased from 10.00 to 4.63 (p < 0.001). Plasma kynurenine concentration correlated with CNS toxicity score: for every 1 µmol/L increase in kynurenine concentration observed, a 1.7 point decrease was observed in CNS toxicity score (p < 0.038). A similar trend was observed for the kynurenine/tryptophan ratio: for every 1 µmol/mmol increase observed in kynurenine/tryptophan ratio, a 0.1 point decrease was observed in CNS toxicity score (p = 0.054). Switching from efavirenz to dolutegravir was associated with increases in plasma kynurenine concentration and improvements in CNS toxicity scores. Underlying mechanisms explaining the rise in kynurenine concentrations need to be established.


Subject(s)
Anti-HIV Agents/therapeutic use , Benzoxazines/toxicity , Central Nervous System/drug effects , HIV Infections/drug therapy , Heterocyclic Compounds, 3-Ring/therapeutic use , Tryptophan/blood , Adult , Alkynes , Antiretroviral Therapy, Highly Active , Benzoxazines/administration & dosage , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Central Nervous System/metabolism , Central Nervous System/pathology , Central Nervous System/virology , Cyclopropanes , Drug Substitution , Female , HIV Infections/blood , HIV Infections/pathology , HIV Infections/virology , Humans , Kynurenine/blood , Male , Middle Aged , Oxazines , Piperazines , Prospective Studies , Pyridones
8.
Drug Metab Dispos ; 47(10): 1195-1205, 2019 10.
Article in English | MEDLINE | ID: mdl-31324697

ABSTRACT

Efavirenz (more specifically the S-enantiomer) is a cornerstone antiretroviral therapy for treatment of HIV infection. The major primary metabolite is S-8-hydroxyefavirenz, which does not have antiretroviral activity but is neurotoxic. Cytochrome P450 2B6 (CYP2B6) is the major enzyme catalyzing S-8-hydroxyefavirenz formation. CYP2B6 genetics and drug interactions are major determinants of clinical efavirenz disposition and dose adjustment. In addition, as a prototypic CYP2B6 substrate, S-efavirenz and analogs can inform on the structure, activity, catalytic mechanisms, and stereoselectivity of CYP2B6. Metabolism of R-efavirenz by CYP2B6 remains unexplored. This investigation assessed S-efavirenz metabolism by clinically relevant CYP2B6 genetic variants. This investigation also evaluated R-efavirenz hydroxylation by wild-type CYP2B6.1 and CYP2B6 variants. S-Efavirenz 8-hydroxylation by wild-type CYP2B6.1 and variants exhibited positive cooperativity and apparent cooperative substrate inhibition. On the basis of Clmax values, relative activities for S-efavirenz 8-hydroxylation were in the order CYP2B6.4 > CYP2B6.1 ≈ CYP2B6.5 ≈ CYP2B6.17 > CYP2B6.6 ≈ CYP2B6.7 ≈ CYP2B6.9 ≈ CYP2B6.19 ≈ CYP2B6.26; CYP2B6.16 and CYP2B6.18 showed minimal activity. Rates of R-efavirenz metabolism were approximately 1/10 those of S-efavirenz for wild-type CYP2B6.1 and variants. On the basis of Clmax values, there was 14-fold enantioselectivity (S > R-efavirenz) for wild-type CYP2B6.1, and 5- to 22-fold differences for other CYP2B6 variants. These results show that both CYP2B6 516G > T (CYP2B6*6 and CYP2B6*9) and 983T > C (CYP2B6*16 and CYP2B6*18) polymorphisms cause canonical diminishment or loss-of-function variants for S-efavirenz 8-hydroxylation, provide a mechanistic basis for known clinical pharmacogenetic differences in efavirenz disposition, and may predict additional clinically important variant alleles. Efavirenz is the most stereoselective CYP2B6 drug substrate yet identified and may be a useful probe for the CYP2B6 active site and catalytic mechanisms. SIGNIFICANCE STATEMENT: Clinical disposition of the antiretroviral S-efavirenz is affected by CYP2B6 polymorphisms. Expressed CYP2B6 with 516G>T (CYP2B6*6 and CYP2B6*9), and 983T>C (CYP2B6*16 and CYP2B6*18) polymorphisms had a diminishment or loss of function for efavirenz 8-hydroxylation. This provides a mechanistic basis for efavirenz clinical pharmacogenetics and may predict additional clinically important variant alleles. Efavirenz metabolism showed both cooperativity and cooperative substrate inhibition. With greater than 10-fold enantioselectivity (S- vs. R- metabolism), efavirenz is the most stereoselective CYP2B6 drug substrate yet identified. These findings may provide mechanistic insights.


Subject(s)
Benzoxazines/metabolism , Benzoxazines/pharmacokinetics , Cytochrome P-450 CYP2B6/genetics , HIV Infections/drug therapy , Reverse Transcriptase Inhibitors/pharmacokinetics , Alkynes , Animals , Benzoxazines/administration & dosage , Benzoxazines/chemistry , Benzoxazines/toxicity , Cell Line , Cyclopropanes , Cytochrome P-450 CYP2B6/metabolism , HIV Infections/genetics , Humans , Insecta , Polymorphism, Single Nucleotide , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/toxicity , Stereoisomerism
9.
Traffic ; 17(12): 1313-1321, 2016 12.
Article in English | MEDLINE | ID: mdl-27621028

ABSTRACT

We have characterized cresyl violet as a membrane-permeant fluorophore that localizes to lysosomes and acidic vacuoles of budding yeast, Drosophila, human, murine and canine cells. An acidotropic weak base, cresyl violet is shown to be virtually insensitive to physiological alkali and divalent cations. Because of its unique spectral properties, it can be used in combination with green, red and far-red fluorophores, is less susceptible to photobleaching than alternative acidotropic probes, and does not undergo photoconversion. At concentrations that yield bright labeling of acidic compartments, cresyl violet does not alter the organellar pH nor does it affect the buffering capacity. Its affordability, together with its chemical and spectral properties, make cresyl violet a superior lysosomal marker devoid of many of the negative characteristics associated with other lysosomal probes.


Subject(s)
Benzoxazines/chemistry , Fluorescent Dyes/chemistry , Lysosomes/chemistry , Animals , Benzoxazines/metabolism , Benzoxazines/toxicity , Dogs , Fluorescent Dyes/metabolism , Fluorescent Dyes/toxicity , HeLa Cells , Humans , Hydrogen-Ion Concentration , Lysosomes/metabolism , Lysosomes/ultrastructure , Madin Darby Canine Kidney Cells , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Spectrometry, Fluorescence
10.
J Neurochem ; 147(1): 71-83, 2018 10.
Article in English | MEDLINE | ID: mdl-29989183

ABSTRACT

Elucidating how cannabinoids affect brain function is instrumental for the development of therapeutic tools aiming to mitigate 'on target' side effects of cannabinoid-based therapies. A single treatment with the cannabinoid receptor agonist, WIN 55,212-2, disrupts recognition memory in mice. Here, we evaluate how prolonged, intermittent (30 days) exposure to WIN 55,212-2 (1 mg/kg) alters recognition memory and impacts on brain metabolism and functional connectivity. We show that chronic, intermittent treatment with WIN 55,212-2 disrupts recognition memory (Novel Object Recognition Test) without affecting locomotion and anxiety-like behaviour (Open Field and Elevated Plus Maze). Through 14 C-2-deoxyglucose functional brain imaging we show that chronic, intermittent WIN 55,212-2 exposure induces hypometabolism in the hippocampal dorsal subiculum and in the mediodorsal nucleus of the thalamus, two brain regions directly involved in recognition memory. In addition, WIN 55,212-2 exposure induces hypometabolism in the habenula with a contrasting hypermetabolism in the globus pallidus. Through the application of the Partial Least Squares Regression (PLSR) algorithm to the brain imaging data, we observed that prolonged WIN 55,212-2 administration alters functional connectivity in brain networks that underlie recognition memory, including that between the hippocampus and prefrontal cortex, the thalamus and prefrontal cortex, and between the hippocampus and the perirhinal cortex. In addition, our results support disturbed lateral habenula and serotonin system functional connectivity following WIN 55,212-2 exposure. Overall, this study provides new insight into the functional mechanisms underlying the impact of chronic cannabinoid exposure on memory and highlights the serotonin system as a particularly vulnerable target.


Subject(s)
Benzoxazines/toxicity , Brain/drug effects , Cannabinoid Receptor Agonists/toxicity , Memory/drug effects , Morpholines/toxicity , Naphthalenes/toxicity , Nerve Net/drug effects , Recognition, Psychology/drug effects , Animals , Globus Pallidus/drug effects , Globus Pallidus/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Maze Learning/drug effects , Mediodorsal Thalamic Nucleus/drug effects , Mediodorsal Thalamic Nucleus/metabolism , Mice , Mice, Inbred C57BL , Neural Pathways/drug effects , Prefrontal Cortex/drug effects
11.
Toxicol Appl Pharmacol ; 339: 34-41, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29198728

ABSTRACT

Flumioxazin, an N-phenylimide herbicide, inhibits protoporphyrinogen oxidase (PPO), a key enzyme in heme biosynthesis in mammals, and causes rat-specific developmental toxicity. The mechanism has mainly been clarified, but no research has yet focused on the contribution of its metabolites. We therefore conducted in vivo metabolism studies in pregnant rats and rabbits, and found 6 major known metabolites in excreta. There was no major rat-specific metabolite. The most abundant component in rat fetuses was APF, followed by flumioxazin and 5 identified metabolites. The concentrations of flumioxazin and these metabolites in fetuses were lower in rabbits than in rats. In vitro PPO inhibition assays with rat and human liver mitochondria showed that flumioxazin is a more potent PPO inhibitor than the metabolites. There were no species differences in relative intensity of PPO inhibition among flumioxazin and these metabolites. Based on the results of these in vivo and in vitro experiments, we concluded that flumioxazin is the causal substance of the rat-specific developmental toxicity. As a more reliable test system for research on in vitro PPO inhibition, cell-based assays with rat, rabbit, monkey, and human hepatocytes were performed. The results were consistent with those of the mitochondrial assays, and rats were more sensitive to PPO inhibition by flumioxazin than humans, while rabbits and monkeys were almost insensitive. From these results, the species difference in the developmental toxicity was concluded to be due to the difference in sensitivity of PPO to flumioxazin, and rats were confirmed to be the most sensitive of these species.


Subject(s)
Benzoxazines/metabolism , Fetal Development/drug effects , Fetus/metabolism , Herbicides/metabolism , Phthalimides/metabolism , Protoporphyrinogen Oxidase/antagonists & inhibitors , Protoporphyrinogen Oxidase/metabolism , Animals , Benzoxazines/toxicity , Female , Fetal Development/physiology , Fetus/drug effects , Haplorhini , Hepatocytes/drug effects , Hepatocytes/metabolism , Herbicides/toxicity , Humans , Phthalimides/toxicity , Pregnancy , Rabbits , Rats , Rats, Sprague-Dawley , Rats, Wistar , Species Specificity
12.
J Cardiovasc Pharmacol ; 72(4): 191-194, 2018 10.
Article in English | MEDLINE | ID: mdl-30080756

ABSTRACT

Cannabinoid CB2 receptor agonists are under investigation for clinical use. At the same time, synthetic cannabinoids have been implicated in a number of deaths. One cause of death is thought to be cardiac arrest subsequent to extreme tachycardia. Central mechanisms are thought to play a role in this, with CB1 but not CB2 receptors thought to mediate central effects. However, the direct effects of cannabinoids on the heart are less well understood. We therefore tested the effects of cannabinoids on isolated rat atria to test whether activation of myocardial CB1 and CB2 receptors could contribute to tachycardia. Although we found a moderate effect that can be attributed to CB1 receptors, we did not find any evidence for chronotropic effects by a CB2 receptor activation. Our results indicate that cannabinoid cardiotoxicity may partially involve CB1 receptors in the myocardium, and that CB2 receptor agonists are unlikely to have significant effects on the heart.


Subject(s)
Arachidonic Acids/pharmacology , Atrial Function/drug effects , Benzoxazines/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Cannabinoids/pharmacology , Heart Atria/drug effects , Morpholines/pharmacology , Naphthalenes/pharmacology , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB2/agonists , Animals , Arachidonic Acids/toxicity , Benzoxazines/toxicity , Cannabinoid Receptor Agonists/toxicity , Cannabinoids/toxicity , Cardiotoxicity , Heart Atria/metabolism , Heart Atria/physiopathology , Heart Rate/drug effects , In Vitro Techniques , Macrophage Activation/drug effects , Male , Mice , Morpholines/toxicity , Myocardial Contraction/drug effects , Naphthalenes/toxicity , RAW 264.7 Cells , Rats, Wistar , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Signal Transduction/drug effects , Tachycardia/chemically induced , Tachycardia/metabolism , Tachycardia/physiopathology
13.
Neurochem Res ; 42(11): 3220-3232, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28770436

ABSTRACT

Although antiretroviral (ARV) therapy has reduced the incidence of severe dementia associated with HIV infection, there has been a rise in milder neurocognitive complaints. Data from HIV patients taking ARVs have shown measurable neurocognitive improvements during drug cessation, suggesting a neurotoxic role of the therapy itself. Mechanisms underlying potential ARV neurotoxicity have not been thoroughly investigated, however pathologic oxidative stress and mitochondrial dysfunction have been suspected. Using DIV 16 primary rat cortical neuron culture, we tested eight ARVs from the three most commonly prescribed ARV classes: nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs/NtRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), and protease inhibitors (PIs) for effects on neuron viability and morphology after 24 h of drug exposure. Of the tested NRTIs, only stavudine at nearly 100 times the target plasma concentration affected neuron viability with no appreciable change in morphology. Dideoxyinosine induced dendritic simplification at 100 times target plasma concentrations, but did not adversely affect viability. The sole NtRTI, tenofovir, induced dendritic simplification at approximately 3-4 times target plasma concentration, but did not affect viability. Of the tested PIs, only amprenavir decreased neuron viability at nearly 100 times the target plasma concentration. The non-nucleoside reverse transcriptase inhibitor, efavirenz, consistently reduced viability (at 50 µM) and induced dendritic simplification (at 20 µM) nearest the target plasma concentration. Probing mitochondrial energetics of DIV16 cortical neurons after exposure to 20 µM efavirenz showed rapid diminution of mitochondrial-dependent oxygen consumption. Further, 20 µM efavirenz decreased excitability in ex vivo slice culture whereas 2 µM had no effect.


Subject(s)
Anti-HIV Agents/toxicity , Anti-Retroviral Agents/toxicity , Benzoxazines/toxicity , Reverse Transcriptase Inhibitors/toxicity , Alkynes , Animals , Animals, Newborn , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Cyclopropanes , Dose-Response Relationship, Drug , Male , Neurons/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley
14.
Cell Biol Toxicol ; 33(1): 69-82, 2017 02.
Article in English | MEDLINE | ID: mdl-27639578

ABSTRACT

Efavirenz is a non-nucleoside reverse transcriptase inhibitor (NNRTI) and an active constituent of the highly active antiretroviral therapy regime. It has significantly contributed in control and management of human immunodeficiency virus propagation. However, EFV administration has also led to severe adverse effects, several reports highlighted the role of EFV in mitochondrial dysfunction and toxicity but the molecular mechanism has been poorly understood. In present study, human hepatoma cells Huh 7.5 were treated with clinically relevant concentrations of EFV and parameters like cytotoxicity, mitochondrial transmembrane potential, mitochondrial morphology, cytochrome c release, mitochondria-mediated apoptosis, mtDNA and mtRNA levels and EFV distribution into mitochondrial compartment were evaluated to understand sequence of events leading to cell death in EFV-treated cells. EFV at its clinically relevant concentration was significantly toxic after 48 and 72 h of treatments. EFV-mediated toxicity is initiated with the permeabilization of mitochondrial outer membrane and change in mitochondrial membrane potential (Δψm) which triggers a series of events like cytochrome c release, alteration in mitochondrial morphology and mitochondria-mediated apoptosis. Total mitochondrial content is reduced after 48 h of EFV treatment at IC50 concentration which is also reflected in reduced mitochondrial DNA and RNA levels. After detecting EFV in mitochondrial compartment after 12 h of incubation with EFV, we hypothesize that EFV being a lipophilic molecule is internalized into the mitochondrial compartment causing depolarization of Δψm which subsequently leads to a cascade of events causing cell death.


Subject(s)
Benzoxazines/toxicity , Membrane Potential, Mitochondrial/drug effects , Reverse Transcriptase Inhibitors/toxicity , Alkynes , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclopropanes , Cytochromes c/metabolism , DNA, Mitochondrial/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Poly(ADP-ribose) Polymerases/metabolism , RNA/metabolism
15.
Neurochem Res ; 41(12): 3278-3288, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27655255

ABSTRACT

Efavirenz is a widely prescribed non-nucleoside reverse transcriptase inhibitor for the treatment of HIV infections. To test for potential long-term consequences of efavirenz on brain cells, cultured primary astrocytes were incubated with this substance or with its primary metabolite 8-hydroxy efavirenz for up to 7 days. Both, efavirenz and 8-hydroxy efavirenz caused time- and concentration-dependent cell toxicity and stimulated in subtoxic concentrations the glycolytic flux (glucose consumption and lactate release) in astrocytes. As 8-hydroxy efavirenz was less toxic than efavirenz and stimulated glycolysis in lower concentrations we tested for a potential hydroxylation of efavirenz to 8-hydroxy efavirenz in astrocytes. Analysis of media and cell lysates by HPLC-UV and mass spectrometry revealed that after 3 days of incubation viable astrocytes had accumulated about 17 and 7 % of the applied efavirenz and 8-hydroxy efavirenz, respectively. However, in cultures treated with efavirenz neither 8-hydroxy efavirenz nor any other known metabolite of efavirenz was detectable. These data demonstrate that cultured rat astrocytes efficiently accumulate, but not metabolize, efavirenz and 8-hydroxy efavirenz and that the observed chronic stimulation of glycolysis is mediated by both efavirenz and 8-hydroxy efavirenz.


Subject(s)
Anti-HIV Agents/toxicity , Astrocytes/drug effects , Benzoxazines/toxicity , Brain/cytology , Reverse Transcriptase Inhibitors/toxicity , Alkynes , Animals , Animals, Newborn , Anti-HIV Agents/chemistry , Anti-HIV Agents/metabolism , Astrocytes/cytology , Astrocytes/metabolism , Benzoxazines/chemistry , Benzoxazines/metabolism , Cell Survival/drug effects , Cells, Cultured , Cyclopropanes , Glucose/metabolism , Lactic Acid/metabolism , Rats, Wistar , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/metabolism , Stereoisomerism
16.
Pharmacol Res ; 110: 10-24, 2016 08.
Article in English | MEDLINE | ID: mdl-27157251

ABSTRACT

Efavirenz is highly effective at suppressing HIV-1, and the WHO guidelines list it as a component of the first-line antiretroviral (ARV) therapies for treatment-naïve patients. Though the pharmacological basis is unclear, efavirenz is commonly associated with a risk for neuropsychiatric adverse events (NPAEs) when taken at the prescribed dose. In many patients these NPAEs appear to subside after several weeks of treatment, though long-term studies show that in some patients the NPAEs persist. In a recent study focusing on the abuse potential of efavirenz, its receptor psychopharmacology was reported to include interactions with a number of established molecular targets for known drugs of abuse, and it displayed a prevailing behavioral profile in rodents resembling an LSD-like activity. In this report, we discovered interactions with additional serotonergic targets that may be associated with efavirenz-induced NPAEs. The most robust interactions were with 5-HT3A and 5-HT6 receptors, with more modest interactions noted for the 5-HT2B receptor and monoamine oxidase A. From a molecular mechanistic perspective, efavirenz acts as a 5-HT6 receptor inverse agonist of Gs-signaling, 5-HT2A and 5-HT2C antagonist of Gq-signaling, and a blocker of the 5-HT3A receptor currents. Efavirenz also completely or partially blocks agonist stimulation of the M1 and M3 muscarinic receptors, respectively. Schild analysis suggests that efavirenz competes for the same site on the 5-HT2A receptor as two known hallucinogenic partial agonists (±)-DOI and LSD. Prolonged exposure to efavirenz reduces 5-HT2A receptor density and responsiveness to 5-HT. Other ARVs such as zidovudine, nevirapine and emtricitabine did not share the same complex pharmacological profile as efavirenz, though some of them weakly interact with the 5-HT6 receptor or modestly block GABAA currents.


Subject(s)
Anti-HIV Agents/toxicity , Benzoxazines/toxicity , Brain/drug effects , HIV Infections/drug therapy , HIV-1/drug effects , Receptors, Serotonin/drug effects , Reverse Transcriptase Inhibitors/toxicity , Serotonin Antagonists/toxicity , Alkynes , Animals , Anti-HIV Agents/metabolism , Benzoxazines/metabolism , Binding, Competitive , Brain/metabolism , CHO Cells , Calcium Signaling/drug effects , Cricetulus , Cyclopropanes , Dose-Response Relationship, Drug , Drug Partial Agonism , Guinea Pigs , HEK293 Cells , HIV Infections/diagnosis , HIV Infections/virology , HIV-1/pathogenicity , HeLa Cells , Humans , Membrane Potentials , Monoamine Oxidase Inhibitors/toxicity , Protein Binding , Radioligand Assay , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/genetics , Receptors, Muscarinic/metabolism , Receptors, Serotonin/genetics , Receptors, Serotonin/metabolism , Reverse Transcriptase Inhibitors/metabolism , Time Factors , Transfection
17.
BMC Infect Dis ; 16: 56, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26831894

ABSTRACT

BACKGROUND: Efavirenz, widely used as part of antiretroviral drug regimens in the treatment of paediatric human immunodeficiency virus infection, has central nervous system side effects. We describe four children presenting with serious, persistent central nervous system adverse events who were found to have elevated plasma efavirenz concentrations as a result of carrying CYP2B6 single nucleotide polymorphisms, known to play a role in the metabolism of EFV. None of the children had a CYP2B6 wildtype haplotype. We believe this is the first case of cerebellar dysfunction associated with efavirenz use to be described in children. CASE PRESENTATION: Four black African children, between the ages of 4 and 8 years presenting between 1 and 20 months post-efavirenz initiation, are described. Cerebellar dysfunction, generalised seizures and absence seizures were the range of presenting abnormalities. Plasma efavirenz levels ranged from 20-60 mg/L, 5-15 times the upper limit of the suggested reference range. All abnormal central nervous system manifestations abated after efavirenz discontinuation. CONCLUSION: Efavirenz toxicity should always be considered in human immunodeficiency virus-infected children with unexplained central nervous system abnormalities. Our findings further our understanding of the impact of genetic variants on antiretroviral pharmacokinetics in children across various ethnic groups. Screening for potential EFV-toxicity based on the CYP2B6 c.516 SNP alone, may not be adequate.


Subject(s)
Anti-HIV Agents/toxicity , Benzoxazines/toxicity , Central Nervous System/drug effects , Cytochrome P-450 CYP2B6/genetics , HIV Infections/drug therapy , Polymorphism, Single Nucleotide , Alkynes , Anti-HIV Agents/blood , Anti-HIV Agents/metabolism , Anti-HIV Agents/therapeutic use , Benzoxazines/blood , Benzoxazines/metabolism , Benzoxazines/therapeutic use , Child , Child, Preschool , Cyclopropanes , Female , Haplotypes , Humans , Male
18.
J Infect Dis ; 211(12): 1953-8, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25538272

ABSTRACT

The anti-human immunodeficiency virus (HIV) drug efavirenz (EFV) alters mitochondrial function in cultured neurons and glial cells. Nitric oxide (NO) is a mediator of mitochondrial dysfunction associated with HIV central nervous system symptoms. We show that EFV promotes inducible nitric oxide synthase (iNOS) expression in cultured glial cells and generated NO undermines their mitochondrial function, as inhibition of NOS partially reverses this effect. EFV inhibits mitochondrial Complex I in both neurons and glia; however, when the latter cells are treated for longer periods, other mitochondrial complexes are also affected in accordance with the increased NO production. These findings shed light on the mechanisms responsible for the frequent EFV-associated neurotoxicity.


Subject(s)
Benzoxazines/toxicity , Mitochondria/metabolism , Neuroglia/metabolism , Neurons/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide/metabolism , Alkynes , Anti-HIV Agents/toxicity , Cell Line , Cyclopropanes , Humans
19.
J Antimicrob Chemother ; 69(11): 2995-3000, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25011651

ABSTRACT

OBJECTIVES: Growing evidence associates the non-nucleoside reverse transcriptase inhibitor efavirenz with several adverse events. Newer antiretrovirals, such as the integrase inhibitor raltegravir, the non-nucleoside reverse transcriptase inhibitor rilpivirine and the protease inhibitor darunavir, claim to have a better toxicological profile than efavirenz while producing similar levels of efficacy and virological suppression. The objective of this study was to determine the in vitro toxicological profile of these three new antiretrovirals by evaluating their effects on the mitochondrial and cellular parameters altered by efavirenz in hepatocytes and neurons. METHODS: Hep3B cells and primary rat neurons were treated with clinically relevant concentrations of efavirenz, darunavir, rilpivirine or raltegravir. Parameters of mitochondrial function, cytotoxicity and oxidative and endoplasmic reticulum stress were assessed using standard cell biology techniques. RESULTS: None of the new compounds altered the mitochondrial function of hepatic cells or neurons, while efavirenz decreased mitochondrial membrane potential and enhanced superoxide production in both cell types, effects that are known to significantly compromise the functioning of mitochondria, cell viability and, ultimately, cell number. Of the four drugs assayed, efavirenz was the only one to alter the protein expression of LC3-II, an indicator of autophagy, and CHOP, a marker of endoplasmic reticulum stress and the unfolded protein response. CONCLUSIONS: Darunavir, rilpivirine and raltegravir do not induce toxic effects on Hep3B cells and primary rat neurons, which suggests a safer hepatic and neurological profile than that of efavirenz.


Subject(s)
Benzoxazines/toxicity , Hepatocytes/drug effects , Mitochondria/drug effects , Nitriles/toxicity , Pyrimidines/toxicity , Pyrrolidinones/toxicity , Sulfonamides/toxicity , Alkynes , Animals , Anti-HIV Agents/toxicity , Cell Line, Tumor , Cells, Cultured , Cyclopropanes , Darunavir , Drug Resistance, Viral/drug effects , Hepatocytes/metabolism , Humans , Mitochondria/metabolism , Neurons/drug effects , Neurons/metabolism , Raltegravir Potassium , Rats , Reverse Transcriptase Inhibitors/toxicity , Rilpivirine
20.
Birth Defects Res B Dev Reprod Toxicol ; 101(2): 162-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24753331

ABSTRACT

BACKGROUND: S-53482 and S-23121 are N-phenylimide herbicides and produced embryolethality, teratogenicity (mainly ventricular septal defects and wavy ribs), and growth retardation in rats in conventional oral developmental toxicity studies. Our objective in this study was to investigate whether the compounds induce developmental toxicity via the dermal route, which is more relevant to occupational exposure, hence better addressing human health risks. METHODS: S-53482 was administered dermally to rats at 30, 100, and 300 mg/kg during organogenesis, and S-23121 was administered at 200, 400, and 800 mg/kg (the maximum applicable dose level). Fetuses were obtained by a Cesarean section and examined for external, visceral, and skeletal alterations. RESULTS: Dermal exposure of rats to S-53482 at 300 mg/kg produced patterns of developmental toxicity similar to those resulting from oral exposure. Toxicity included embryolethality, teratogenicity, and growth retardation. Dermal administration of S-23121 at 800 mg/kg resulted in an increased incidence of embryonic death and ventricular septal defect, but retarded fetal growth was not observed as it was following oral exposure to S-23121. CONCLUSIONS: Based on the results, S-53482 and S-23121 were teratogenic when administered dermally to pregnant rats as were the compounds administered orally. Thus, investigation of the mechanism and its human relevancy become more important.


Subject(s)
Benzoxazines/toxicity , Fetus/drug effects , Herbicides/toxicity , Phthalimides/toxicity , Administration, Cutaneous , Animals , Dose-Response Relationship, Drug , Embryo Loss/chemically induced , Female , Fetus/embryology , Fetus/pathology , Male , Maternal Exposure/adverse effects , Pregnancy , Rats , Rats, Sprague-Dawley , Teratogens/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL