Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 133
1.
Brain Res Bull ; 180: 24-30, 2022 03.
Article En | MEDLINE | ID: mdl-34990732

The main pathological changes that occur in delayed encephalopathy after acute carbon monoxide poisoning (DEACMP) are extensive demyelination of brain white matter and neuron damage. Previous studies suggested that demyelination and neuron injury are related to activating the Rho/ROCK signaling pathway. Inhibition of the Rho/ROCK signaling pathway can alleviate neuron injury and promote myelin repair. This study utilized a DEACMP model in which rats were prepared by space injection of CO gas intraperitoneally (CO group), and the association between the Rho/ROCK signaling pathway and DEACMP was investigated. The ROCK2 kinase inhibitor Y-27632 was used to prevent the effects of the DEACMP model to elucidate its protective mechanism. The results demonstrated that the cognitive and motor functions were significantly impaired, and the GFAP, NSE, RhoA, and ROCK2 protein levels were significantly increased in the CO group within three weeks after the model was established. After Y-27632 intervention, the cognitive and motor functions of the CO+Y-27632 group were significantly improved within three weeks after the model was established. In the CO+Y-27632 group, the RhoA, ROCK2, GFAP, and NSE (indicating neuron injury) protein levels decreased significantly, and the MBP protein levels (indicating myelin repair) increased significantly within three weeks after the model was established. These results suggested that the pathogenesis of DEACMP was associated with activation of the Rho/ROCK pathway and that Y-27632 inhibited ROCK2 kinase activity in the CO exposed rats, resulting in improved behavioral deficits, reduced neuron damage, and promotion of myelin repair. Therefore, Y-27632 might be a potentially effective drug for the treatment of DEACMP-induced brain damage.


Behavior, Animal/drug effects , Carbon Monoxide Poisoning/drug therapy , Cognitive Dysfunction/drug therapy , Neurotoxicity Syndromes/drug therapy , Protein Kinase Inhibitors/pharmacology , rho-Associated Kinases/metabolism , Amides/pharmacology , Animals , Carbon Monoxide Poisoning/complications , Carbon Monoxide Poisoning/enzymology , Cognitive Dysfunction/etiology , Disease Models, Animal , Neurotoxicity Syndromes/complications , Neurotoxicity Syndromes/enzymology , Pyridines/pharmacology , Rats , Signal Transduction/drug effects , rho-Associated Kinases/antagonists & inhibitors
2.
Toxicology ; 465: 153052, 2022 01 15.
Article En | MEDLINE | ID: mdl-34838597

Bisphenol-A (BPA), an environmental endocrine disruptor, is toxic to the central nervous system. Although recent studies have shown BPA-induced neurotoxicity, it is far from clear what precisely epigenetic mechanisms are involved in BPA-induced cognitive deficits. In this study, pheochromocytoma (PC12) cells were treated with BPA at 1 µM for 36 h in vitro. In vivo, C57BL/6 mice were administered to BPA at a dose of 1 mg/kg/day for 10 weeks. The results showed that 1 µM BPA exposure for 36 h impaired neurite outgrowth of PC12 cells through decreasing the primary and secondary branches. Besides, BPA exposure decreased the level of Ac-H3K9 (histone H3 Lys9 acetylation) by upregulating the expression of HDAC2 (histone deacetylases 2) in PC12 cells. Furthermore, treatment of both TSA (Trichostatin A, inhibitor of the histone deacetylase) and shHDAC2 plasmid (HDAC2 knockdown construct) resulted in amelioration neurite outgrowth deficits induced by BPA. In addition, it was shown that repression of HDAC2 could markedly rescue the spine density impairment in the hippocampus and prevent the cognitive impairment caused by BPA exposure in mice. Collectively, HDAC2 plays an essential role in BPA-induced neurotoxicity, which provides a potential molecular target for medical intervention.


Benzhydryl Compounds/toxicity , Dendritic Spines/drug effects , Environmental Pollutants/toxicity , Hippocampus/drug effects , Histone Deacetylase 2/metabolism , Neurites/drug effects , Neurotoxicity Syndromes/etiology , Phenols/toxicity , Animals , Behavior, Animal/drug effects , Cognition/drug effects , Dendritic Spines/enzymology , Dendritic Spines/pathology , Female , Hippocampus/enzymology , Hippocampus/pathology , Hippocampus/physiopathology , Histone Deacetylase 2/genetics , Male , Maze Learning/drug effects , Mice, Inbred C57BL , Neurites/enzymology , Neurites/pathology , Neuronal Outgrowth/drug effects , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/physiopathology , PC12 Cells , Rats , Up-Regulation
3.
Cell Rep ; 37(3): 109872, 2021 10 19.
Article En | MEDLINE | ID: mdl-34686345

SARM1 is an inducible TIR-domain NAD+ hydrolase that mediates pathological axon degeneration. SARM1 is activated by an increased ratio of NMN to NAD+, which competes for binding to an allosteric activating site. When NMN binds, the TIR domain is released from autoinhibition, activating its NAD+ hydrolase activity. The discovery of this allosteric activating site led us to hypothesize that other NAD+-related metabolites might activate SARM1. Here, we show the nicotinamide analog 3-acetylpyridine (3-AP), first identified as a neurotoxin in the 1940s, is converted to 3-APMN, which activates SARM1 and induces SARM1-dependent NAD+ depletion, axon degeneration, and neuronal death. In mice, systemic treatment with 3-AP causes rapid SARM1-dependent death, while local application to the peripheral nerve induces SARM1-dependent axon degeneration. We identify 2-aminopyridine as another SARM1-dependent neurotoxin. These findings identify SARM1 as a candidate mediator of environmental neurotoxicity and suggest that SARM1 agonists could be developed into selective agents for neurolytic therapy.


Armadillo Domain Proteins/metabolism , Axons/drug effects , Cytoskeletal Proteins/metabolism , Ganglia, Spinal/drug effects , Nerve Degeneration , Neurotoxicity Syndromes/etiology , Neurotoxins/toxicity , Pyridines/toxicity , Sciatic Nerve/drug effects , Activation, Metabolic , Allosteric Regulation , Animals , Armadillo Domain Proteins/genetics , Axons/enzymology , Axons/pathology , Catalytic Domain , Cell Death , Cytokines/genetics , Cytokines/metabolism , Cytoskeletal Proteins/genetics , Enzyme Activation , Female , Ganglia, Spinal/enzymology , Ganglia, Spinal/pathology , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/pathology , Neurotoxins/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , Nicotinamide Phosphoribosyltransferase/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Pyridines/metabolism , Sciatic Nerve/enzymology , Sciatic Nerve/pathology , Signal Transduction
4.
Toxicol Appl Pharmacol ; 430: 115723, 2021 11 01.
Article En | MEDLINE | ID: mdl-34520793

Pyrethroids are one of the most commonly used classes of synthetic pesticides in the world. Recent laboratory and epidemiological evidence suggested that pyrethroids have potential adverse effects in the mammalian brain; however, the underlying mechanisms of the neurotoxic effects of pyrethroids have not been fully elucidated. In the present study, we investigated the mechanisms of effects of a type II pyrethroid deltamethrin (DM) in a neuronal cell model focusing on the proteolytic function, including autophagy and the ubiquitin-proteasome system. We confirmed that a micromolar concentration of DM dose-dependently decreased the cell viability and induced apoptotic cell death. Our results showed that DM enhanced autophagy in association with an accumulation of autophagosomes and increase in the levels of autophagy markers LC3-II/LC3-I ratio and p62 which were much elevated in the presence of lysosomal inhibitors bafilomycin A1 and chloroquine. We also found that DM caused a dysfunction of mitochondria with a decrease of mitochondrial membrane potential and mitochondrial DNA copy number as well as colocalization with autophagosomes. Moreover, a decrease in the activities of three major proteasomal enzymes and an accumulation of ubiquitinated proteins were observed by the exposure to DM. Transcriptome analysis revealed that up-regulated genes supported the activation of autophagy with induction of cellular stress responses including oxidative stress and endoplasmic reticulum stress, while down-regulated genes related to the cell cycle and DNA replication. These findings provide novel insights into the neurotoxicity of DM which underlie the imbalance in proteolytic function caused by mitophagy activation and proteasome inhibition.


Insecticides/toxicity , Mitochondria/drug effects , Mitophagy/drug effects , Neurons/drug effects , Neurotoxicity Syndromes/etiology , Nitriles/toxicity , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/toxicity , Pyrethrins/toxicity , Animals , Apoptosis/drug effects , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation , Mice , Mitochondria/enzymology , Mitochondria/genetics , Mitochondria/pathology , Neurons/enzymology , Neurons/pathology , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/pathology , Oxidative Stress/drug effects , Proteolysis , Transcriptome
5.
J Biochem Mol Toxicol ; 35(9): e22855, 2021 Sep.
Article En | MEDLINE | ID: mdl-34309959

Valproic acid (VPA) is an efficient antiepileptic drug widely used for the treatment of epilepsy and other seizures in both children and adults. It is also reported to have side and toxic effects on many organs and tissues. Vitamin B6 (Vit B6 ) is a well-described water-soluble vitamin, which has an antioxidant effect. In this study, we aimed to investigate the protective effect of Vit B6 on VPA-induced brain injury. Male Sprague-Dawley rats were divided into four groups. Group I, control animals; Group II, Vit B6 (50 mg/kg/day) given rats; Group III, VPA (500 mg/kg/day) given rats; Group IV, VPA and Vit B6 given rats at same dose and time. VPA and Vit B6 were administered intraperitoneally and orally, respectively, for 7 days. At the end of the experiments, the rats were sacrificed and brain tissues were taken. Protein carbonyl and sialic acid levels, xanthine oxidase, adenosine deaminase, acetylcholine esterase, lactate dehydrogenase, myeloperoxidase activities, total oxidant status, and reactive oxygen species levels were found to be increased, while glutathione and total antioxidant capacity levels, catalase, superoxide dismutase, glutathione-S-transferase, paraoxonase, and glutathione reductase activities were found to be decreased in the VPA group. Administration of Vit B6 reversed these defects in the VPA group. These findings indicate that Vit B6 has a protective effect on VPA-induced brain damage.


Brain Injuries , Neurotoxicity Syndromes , Valproic Acid/adverse effects , Vitamin B 6/pharmacology , Animals , Brain Injuries/chemically induced , Brain Injuries/drug therapy , Brain Injuries/enzymology , Male , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/enzymology , Oxidoreductases/metabolism , Rats , Rats, Sprague-Dawley , Valproic Acid/pharmacology
6.
Toxicol Lett ; 347: 36-44, 2021 Sep 01.
Article En | MEDLINE | ID: mdl-33945864

Aconitine, a highly toxic alkaloid derived from Aconitum L., affects the central nervous system and peripheral nervous system. However, the underlying mechanism of aconitine-induced neurotoxicity remains unclear. This study investigates the effects and mechanism of aconitine on mitochondrial energy metabolism in SH-SY5Y cells. Results demonstrated that aconitine exposure suppressed cell proliferation and led to an increase in reactive oxygen species (ROS) and excessive lactate dehydrogenase (LDH) release. Aconitine (400 µmol/L) induced abnormal mitochondrial energy metabolism that quantified by the significant decrease in ATP production, basal respiration, proton leak, maximal respiration, and succinate dehydrogenase (SDH) activity. Phosphorylation of AMPK was significantly reduced in aconitine-treated SH-SY5Y cells. The AMPK activator AIACR pretreatment effectively promoted ATP production to ameliorate mitochondrial energy metabolism disorder caused by aconitine. Mitochondrial biosynthesis was inhibited after treatment with 400 µmol/L aconitine, which was characterized by mitochondria number, TFAM expression, and mtDNA copy number. Moreover, aconitine prompted the down-regulation of mitochondrial fusion proteins OPA1, Mfn1 and Mfn2, and the up-regulation of mitochondrial fission proteins p-Drp1 and p-Mff. These results suggest that aconitine induces mitochondrial energy metabolism dysfunction in SH-SY5Y cells, which may involve the inhibition of AMPK signaling and abnormal mitochondrial dynamics.


AMP-Activated Protein Kinases/metabolism , Aconitine/toxicity , Energy Metabolism/drug effects , Mitochondria/drug effects , Mitochondrial Dynamics/drug effects , Neurons/drug effects , Neurotoxicity Syndromes/etiology , Adenosine Triphosphate/metabolism , Cell Line, Tumor , Humans , Mitochondria/enzymology , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Neurons/enzymology , Neurons/pathology , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/pathology , Phosphorylation , Signal Transduction
7.
J Biochem Mol Toxicol ; 35(5): e22735, 2021 May.
Article En | MEDLINE | ID: mdl-33522075

The neurotoxic level of ammonia in the brain during liver cirrhosis causes a nervous system disorder, hepatic encephalopathy (HE), by affecting mitochondrial functions. Sirtuin-3 (SIRT3) is emerging as a master regulator of mitochondrial integrity, which is currently being focused as a pathogenic hotspot for HE. This article describes SIRT3 level versus mitochondrial dysfunction markers in the hippocampus of the control, the moderate-grade hepatic encephalopathy (MoHE), developed in thioacetamide-induced (100 mg/kg bw ip for 10 days) liver cirrhotic rats, and the MoHE rats treated with an SIRT3 activator, honokiol (HKL; 10 mg/kg bw ip), for 7 days from 8th day of the thioacetamide schedule. As compared with the control group rats, hippocampus mitochondria of MoHE rats showed a significant decline in SIRT3 expression and its activity with concordant enhancement of ROS and declined membrane permeability transition and organelle viability scores. This was consistent with the declined mitochondrial thiol level and thiol-regenerating enzyme, isocitrate dehydrogenase 2. Also, significantly declined activities of electron transport chain complexes I, III, IV, and Q10 , decreased NAD+ /NADH and ATP/AMP ratios, and enhanced number of the shrunken mitochondria were recorded in the hippocampus of those MoHE rats. However, all these mitochondrial aberrations were observed to regain their normal profiles/levels, concordant to the enhanced SIRT3 expression and its activity due to treatment with HKL. The findings suggest a role of SIRT3 in mitochondrial structure-function derangements associated with MoHE pathogenesis and SIRT3 activation by HKL as a relevant strategy to protect mitochondrial integrity during ammonia neurotoxicity.


Ammonia/metabolism , Hepatic Encephalopathy/enzymology , Hippocampus/enzymology , Mitochondria/enzymology , Neurotoxicity Syndromes/enzymology , Sirtuins/metabolism , Animals , Disease Models, Animal , Hepatic Encephalopathy/pathology , Hippocampus/pathology , Male , Mitochondria/pathology , Neurotoxicity Syndromes/pathology , Rats
8.
J Biochem Mol Toxicol ; 35(4): e22694, 2021 Apr.
Article En | MEDLINE | ID: mdl-33393683

Alzheimer's disease (AD) is a major public health crisis due to devastating cognitive symptoms, a lack of curative treatments, and increasing prevalence. Most cases are sporadic (>95% of cases) after the age of 65 years, implicating an important role of environmental factors in disease pathogenesis. Environmental neurotoxicants have been implicated in neurodegenerative disorders including Parkinson's Disease and AD. Animal models of AD and in vitro studies have shed light on potential neuropathological mechanisms, yet the biochemical and molecular underpinnings of AD-relevant environmental neurotoxicity remain poorly understood. Beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) is a potentially critical pathogenic target of environmentally induced neurotoxicity. BACE1 clearly has a critical role in AD pathophysiology: It is required for amyloid beta production and expression and activity of BACE1 are increased in the AD brain. Though the literature on BACE1 in response to environmental insults is limited, current studies, along with extensive AD neurobiology literature suggest that BACE1 deserves attention as an important neurotoxic target. Here, we critically review research on environmental neurotoxicants such as metals, pesticides, herbicides, fungicides, polyfluoroalkyl substances, heterocyclic aromatic amines, advanced glycation end products, and acrolein that modulate BACE1 and potential mechanisms of action. Though more research is needed to clearly understand whether BACE1 is a critical mediator of AD-relevant neurotoxicity, available reports provide convincing evidence that BACE1 is altered by environmental risk factors associated with AD pathology, implying that BACE1 inhibition and its use as a biomarker should be considered in AD management and research.


Alzheimer Disease , Amyloid Precursor Protein Secretases/biosynthesis , Aspartic Acid Endopeptidases/biosynthesis , Brain/enzymology , Environmental Exposure/adverse effects , Gene Expression Regulation, Enzymologic , Neurotoxicity Syndromes , Neurotoxins/toxicity , Aged , Alzheimer Disease/enzymology , Alzheimer Disease/etiology , Alzheimer Disease/pathology , Animals , Brain/pathology , Disease Models, Animal , Female , Humans , Male , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/pathology
9.
J Steroid Biochem Mol Biol ; 205: 105776, 2021 01.
Article En | MEDLINE | ID: mdl-33130020

Solasodine analogues containing a seven-membered F ring with a nitrogen atom placed at position 22a were prepared from diosgenin or tigogenin in a four-step synthesis comprising of the simultaneous opening of the F-ring and introduction of cyanide in position 22α, activation of the 26-hydroxyl group as mesylate, nitrile reduction, and N-cyclization. Solasodine, six obtained 22a(N)-homo analogues, as well as four 26a-homosolasodine derivatives and their open-chain precursors (13 in total) were tested as potential inhibitors of acetyl- and butyryl-cholinesterases and showed activity at micromolar concentrations. The structure-activity relationship study revealed that activities against studied esterases are affected by the structure of E/F rings and the substitution pattern of ring A. The most potent compound 8 acted as non-competitive inhibitors and exerted IC50 = 8.51 µM and 7.05 µM for eeAChE and eqBChE, respectively. Molecular docking studies revealed the hydrogen bond interaction of 8 with S293 of AChE; further rings are stabilized via hydrophobic interaction (ring A) or interaction with Y341 and W286 (rings B and C). Biological experiments showed no neurotoxicity of differentiated SH-SY5Y cells. More importantly, results from neuroprotective assay based on glutamate-induced cytotoxicity revealed that most derivatives had the ability to increase the viability of differentiated SH-SY5Y cells in comparison to galantamine and lipoic acid assayed as standards. The newly synthesized solasodine analogues are able to inhibit and to bind cholinesterases in noncompetitive mode of inhibition and exhibited neuroprotection potential of differentiated neuroblastoma cells after Glu-induced toxicity.


Cholinesterase Inhibitors/chemistry , Neuroprotective Agents/chemistry , Neurotoxicity Syndromes/drug therapy , Solanaceous Alkaloids/chemistry , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/pharmacology , Cholinesterases/chemistry , Cholinesterases/drug effects , Diosgenin/chemistry , Humans , Molecular Docking Simulation , Molecular Structure , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/pathology , Nitrogen/chemistry , Solanaceous Alkaloids/chemical synthesis , Solanaceous Alkaloids/pharmacology , Structure-Activity Relationship
10.
Mol Neurobiol ; 57(11): 4608-4621, 2020 Nov.
Article En | MEDLINE | ID: mdl-32761352

CYP2D6 metabolically inactivates several neurotoxins, including beta-carbolines, which are implicated in neurodegenerative diseases. Variation in CYP2D6 within the brain may alter local inactivation of neurotoxic beta-carbolines, thereby influencing neurotoxicity. The beta-carboline harmine, which induces hypothermia and tremor, is metabolized by CYP2D6 to the non-hypothermic/non-tremorgenic harmol. Transgenic mice (TG), expressing human CYP2D6 in addition to their endogenous mouse CYP2D, experience less harmine-induced hypothermia and tremor compared with wild-type mice (WT). We first sought to elucidate the role of CYP2D in general within the brain in harmine-induced hypothermia and tremor severity. A 4-h intracerebroventricular (ICV) pretreatment with the CYP2D inhibitor propranolol increased harmine-induced hypothermia and tremor in TG and increased harmine-induced hypothermia in WT. We next sought to specifically demonstrate that human CYP2D6 expressed in TG brain altered harmine response severity. A 24-h ICV propranolol pretreatment, which selectively and irreversibly inhibits human CYP2D6 in TG brain, increased harmine-induced hypothermia. This 24-h pretreatment had no impact on harmine response in WT, as propranolol is not an irreversible inhibitor of mouse CYP2D in the brain, thus confirming no off-target effects of ICV propranolol pretreatment. Human CYP2D6 activity in TG brain was sufficient in vivo to mitigate harmine-induced neurotoxicity. These findings suggest that human CYP2D6 in the brain is protective against beta-carboline-induced neurotoxicity and that the extensive interindividual variability in CYP2D6 expression in human brain may contribute to variation in susceptibility to certain neurotoxin-associated neurodegenerative disorders.


Brain/pathology , Cytochrome P-450 CYP2D6/metabolism , Harmine/toxicity , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/prevention & control , Animals , Brain/drug effects , Brain/enzymology , Humans , Hypothermia, Induced , Injections, Intraventricular , Liver/drug effects , Liver/enzymology , Liver/pathology , Mice, Transgenic , Neurotoxicity Syndromes/complications , Propranolol/administration & dosage , Tremor/complications
11.
Naunyn Schmiedebergs Arch Pharmacol ; 393(9): 1635-1648, 2020 09.
Article En | MEDLINE | ID: mdl-32377769

Pregabalin abuse has become an emerging concern; thus, the current study has been designed to study the neurotoxic hazards of prolonged high-dose of pregabalin (akin to that abused by addicts) and to evaluate the effect of alpha tocopherol as a possible ameliorating agent. The current study evaluated the brain neurotransmitters; dopamine, glutamate, and norepinephrine. The study also assessed the expression of the apoptosis-related markers Bax, Bcl2, and caspase 3. Western-blotted analysis of the three major mitogen-activated protein kinases (MAPKs), the c-JUN N-terminal kinase (JNK), the p38 MAPK, and the extracellular signal-regulated kinase (ERK), has also been performed. The study also evaluated oxidative stress via assessment of the cortical tissue levels of reduced glutathione and malondialdehyde and the activity of superoxide dismutase. Histopathological examination and histomorphometric evaluation of the darkly degenerated cortical neurons have also been performed. Pregabalin in high doses (150 mg/kg/day and 300 mg/kg/day) disrupted the ERK/JNK/p38-MAPK signaling, reversed the bax/bcl2 ratio, and induced oxidative stress. It also diminished the release of dopamine, glutamate, and norepinephrine and increased the count of degenerated neurons. Alpha tocopherol treatment significantly attenuated the deleterious effects induced by pregabalin. The role of alpha tocopherol in ameliorating the oxidative stress injury, and apoptosis induced by pregabalin, along with its role in normalizing neurotransmitters, modulating the ERK/JNK/p38-MAPK signaling pathways and improving the histopathological cortical changes, offers alpha tocopherol as a promising adjunctive therapy in patients undergoing prolonged pregabalin therapy as those suffering from prolonged seizures and neuropathies.


Antioxidants/pharmacology , Apoptosis/drug effects , Cerebral Cortex/drug effects , Mitogen-Activated Protein Kinases/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/prevention & control , Oxidative Stress/drug effects , Pregabalin/toxicity , alpha-Tocopherol/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Male , Neurons/enzymology , Neurons/pathology , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/pathology , Phosphorylation , Rats, Wistar , Signal Transduction
12.
Arch Toxicol ; 94(6): 2149-2162, 2020 06.
Article En | MEDLINE | ID: mdl-32303805

Organophosphate (OP) threat agents can trigger seizures that progress to status epilepticus, resulting in persistent neuropathology and cognitive deficits in humans and preclinical models. However, it remains unclear whether patients who do not show overt seizure behavior develop neurological consequences. Therefore, this study compared two subpopulations of rats with a low versus high seizure response to diisopropylfluorophosphate (DFP) to evaluate whether acute OP intoxication causes persistent neuropathology in non-seizing individuals. Adult male Sprague Dawley rats administered DFP (4 mg/kg, sc), atropine sulfate (2 mg/kg, im), and pralidoxime (25 mg/kg, im) were monitored for seizure activity for 4 h post-exposure. Animals were separated into groups with low versus high seizure response based on behavioral criteria and electroencephalogram (EEG) recordings. Cholinesterase activity was evaluated by Ellman assay, and neuropathology was evaluated at 1, 2, 4, and 60 days post-exposure by Fluoro-Jade C (FJC) staining and micro-CT imaging. DFP significantly inhibited cholinesterase activity in the cortex, hippocampus, and amygdala to the same extent in low and high responders. FJC staining revealed significant neurodegeneration in DFP low responders albeit this response was delayed, less persistent, and decreased in magnitude compared to DFP high responders. Micro-CT scans at 60 days revealed extensive mineralization that was not significantly different between low versus high DFP responders. These findings highlight the importance of considering non-seizing patients for medical care in the event of acute OP intoxication. They also suggest that OP intoxication may induce neurological damage via seizure-independent mechanisms, which if identified, might provide insight into novel therapeutic targets.


Brain Waves/drug effects , Brain/drug effects , Cholinesterase Inhibitors/toxicity , Convulsants/toxicity , Isoflurophate/toxicity , Nerve Degeneration , Neurotoxicity Syndromes/etiology , Seizures/chemically induced , Acetylcholinesterase/metabolism , Animals , Behavior, Animal/drug effects , Brain/diagnostic imaging , Brain/enzymology , Brain/physiopathology , GPI-Linked Proteins/metabolism , Male , Neurotoxicity Syndromes/diagnostic imaging , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/physiopathology , Rats, Sprague-Dawley , Seizures/diagnostic imaging , Seizures/enzymology , Seizures/physiopathology , Time Factors , X-Ray Microtomography
13.
Toxicology ; 438: 152442, 2020 05 30.
Article En | MEDLINE | ID: mdl-32278051

Bisphenol A (BPA) is a well-known endocrine disruptor used to manufacture polycarbonate plastics and epoxy resins. BPA exposure especially occupational perinatal exposure to has been linked to numerous adverse effects for the offspring. Available data have shown that perinatal exposure to BPA contributes to neurodegenerative pathological changes; however, the potential mechanisms remain unclear. This study attempted to investigate the long-term consequences of perinatal exposure to BPA on the offspring mouse brain. The pregnant mice were given either a vehicle control or BPA (2, 10, 100 µg/kg/d) from day 6 of gestation until weaning (P6-PND21, foetal and neonatal exposure). At 3, 6 and 9 months of age, the neurotoxic effects in the offspring in each group were investigated. We found that the spine density but not the dendritic branches in the hippocampus were noticeably reduced at 6 and 9 months of age. Meanwhile, p-Tau, the characteristic protein for tauopathy, was dramatically increased in both the hippocampus and cortex at 3-9 months of age. Mechanically, the balance of kinase and protein phosphatase, which plays critical roles in p-Tau regulation, was disturbed. It indicated that GSK3ß and CDK5, two critical kinases, were activated in most of the BPA perinatal exposure group, while protein phosphatase 2A (PP2A), one of the important phosphatases, regulated p-Tau expression through its demethylation, methylation and phosphorylation. Taken together, the present study may be translatable to the human occupational BPA exposure due to a similar exposure level. BPA perinatal exposure causes long-term adverse effects on the mouse brain and may be a risk factor for tauopathies, and the CDK5/GSK3ß/PP2A axis might be a promising therapeutic target for BPA-induced neurodegenerative pathological changes.


Benzhydryl Compounds/toxicity , Cerebral Cortex/drug effects , Cyclin-Dependent Kinase 5/metabolism , Endocrine Disruptors/toxicity , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/drug effects , Neurotoxicity Syndromes/etiology , Phenols/toxicity , Prenatal Exposure Delayed Effects , Protein Phosphatase 2/metabolism , tau Proteins/metabolism , Animals , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Dendritic Spines/drug effects , Dendritic Spines/enzymology , Dendritic Spines/pathology , Female , Gestational Age , Hippocampus/enzymology , Hippocampus/pathology , Male , Maternal Exposure , Mice, Inbred C57BL , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/pathology , Phosphorylation , Pregnancy
14.
Biomolecules ; 10(3)2020 03 09.
Article En | MEDLINE | ID: mdl-32182821

Exposure of neural cells to harmful and toxic factors promotes oxidative stress, resulting in disorders of metabolism, cell differentiation, and maturation. The study examined the brains of rats pre- and postnatally exposed to sodium fluoride (NaF 50 mg/L) and activity of NADPH oxidase 4 (NOX4), catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPx), glutathione reductase (GR), concentration of glutathione (GSH), and total antioxidant capacity (TAC) in the cerebellum, prefrontal cortex, hippocampus, and striatum were measured. Additionally, NOX4 expression was determined by qRT-PCR. Rats exposed to fluorides (F-) showed an increase in NOX4 activity in the cerebellum and hippocampus, a decrease in its activity in the prefrontal cortex and hippocampus, and upregulation of NOX4 expression in hippocampus and its downregulation in other brain structures. Analysis also showed significant changes in the activity of all antioxidant enzymes and a decrease in TAC in brain structures. NOX4 induction and decreased antioxidant activity in central nervous system (CNS) cells may be central mechanisms of fluoride neurotoxicity. NOX4 contributes to blood-brain barrier damage, microglial activation, and neuronal loss, leading to impairment of brain function. Fluoride-induced oxidative stress involves increased reactive oxygen speciaes (ROS) production, which in turn increases the expression of genes encoding pro-inflammatory cytokines.


Brain/enzymology , Gene Expression Regulation, Enzymologic/drug effects , Glutathione/metabolism , NADPH Oxidase 4/biosynthesis , Neurotoxicity Syndromes/enzymology , Prenatal Exposure Delayed Effects/enzymology , Sodium Fluoride/toxicity , Up-Regulation/drug effects , Animals , Disease Models, Animal , Female , Oxidative Stress/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
15.
Toxicol Lett ; 322: 66-76, 2020 Apr 01.
Article En | MEDLINE | ID: mdl-31945382

Silent Information Regulator 1 (SIRT1), an NAD+-dependent deacetylase, contributes to the neuroprotective effect. However, intracellular signaling pathways that affect SIRT1 function remain unknown. It is well known that N-methyl-D-aspartate (NMDA) receptor activation induces calcium influx which then activates PKC, and SIRT1 is a mRNA target for HuR protein. We hypothesize that Ca2+-PKC-HuR-SIRT1 pathway modulates SIRT1 function. The present study is to investigate the potential pathway of SIRT1 in the SH-SY5Y cell line as an in vitro model of NMDA-induced neurotoxicity. The results showed that: (1) SIRT1 levels were downregulated in NMDA model; (2) NMDA induced an increase in serine phosphorylation of HuR, while inhibition of serine phosphorylation of HuR increased SIRT1 levels, promoting cell survival; (3) PKC inhibitor (Gö 6976) reversed NMDA insults and also suppressed serine phosphorylation of HuR; (4) 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA-AM), an intracellular calcium chelator, fully reversed NMDA insults and also inhibited PKC activity evoked by NMDA. These results indicate that intracellular elevated Ca2+ activates PKC, which phosphorylates HuR and then promotes SIRT1 mRNA decay and subsequent neuronal death in NMDA model. Therefore, the study suggests that inhibition of Ca2+-PKC-HuR-SIRT1 pathway could be an effective strategy for preventing certain neurological diseases related to NMDA excitotoxicity.


Excitatory Amino Acid Agonists/toxicity , N-Methylaspartate/toxicity , Neurons/drug effects , Neurotoxicity Syndromes/etiology , Sirtuin 1/metabolism , Calcium/metabolism , Calcium Signaling , Cell Death/drug effects , Cell Line, Tumor , Down-Regulation , ELAV-Like Protein 1/metabolism , Humans , Neurons/enzymology , Neurons/pathology , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/pathology , Phosphorylation , Protein Kinase C/metabolism , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Serine , Sirtuin 1/genetics
16.
Toxicol Lett ; 318: 104-113, 2020 Jan.
Article En | MEDLINE | ID: mdl-31672611

BACKGROUND: Bupivacaine (BP) is commonly used as a local anaesthetic(LA) in the clinic, but it can also cause neurotoxicity, especially in patients with diabetes. Previous studies have found that high-glucose environments can aggravate BP-induced DNA damage in nerve cells. Ku70 is subunit of the DNA damage repair enzyme DNA-PK. This study was designed to determine whether high-glucose conditions enhance BP neurotoxicity and DNA damage by inhibiting Ku70 expression. METHODS: We examined the effect of BP on apoptosis and DNA damage in murine dorsal root ganglion (DRG) neurons under hyperglycaemic conditions. Untreated DRG cells and DRG cells pretreated with NU7441, a DNA-PK inhibitor, were cultured for 3 days under normal culture conditions or with 50 mM glucose, and the cells were then treated with BP for 3 h. DNA damage was investigated via comet assays, the ratio of early to late apoptotic cells was assessed by Annexin V-FITC/PI staining, and cell viability was measured by CCK-8 assays. The protein expression levels of DNA-PK, Ku70, Bax, Bcl-2 and γH2ax were measured by immunofluorescence or Western blotting. RESULTS: Compared to its effect under normal culture conditions, BP treatment led to decreased cell viability and increased DNA damage in DRG cells grown under high-glucose conditions. The rate of DRG cell apoptosis and the expression of γH2ax, the ratio of Bax to Bcl-2 also increased under the high-glucose conditions. Furthermore, Ku70 expression was inhibited. The DNA-PK inhibitor, NU7441, could significantly inhibit DNA-PK and Ku70 expression, simultaneously further aggravating BP-induced apoptosis and DNA damage under high-glucose conditions. CONCLUSION: These data indicate that hyperglycaemia may enhance BP-induced neurotoxicity and DNA damage by inhibiting the DNA repair protein Ku70.


Anesthetics, Local/toxicity , Apoptosis/drug effects , Bupivacaine/toxicity , Chromones/toxicity , Enzyme Inhibitors/toxicity , Ganglia, Spinal/drug effects , Glucose/toxicity , Ku Autoantigen/antagonists & inhibitors , Morpholines/toxicity , Neurotoxicity Syndromes/etiology , Animals , Cells, Cultured , DNA Damage , Ganglia, Spinal/enzymology , Ganglia, Spinal/pathology , Ku Autoantigen/metabolism , Mice , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/pathology , Signal Transduction/drug effects
17.
Arch Toxicol ; 93(9): 2661-2671, 2019 09.
Article En | MEDLINE | ID: mdl-31332466

Bisphenol analogues including bisphenol A and its derivatives are ubiquitous environmental contaminants and have been linked to adverse neurodevelopment effects on animals and humans. Most toxicological research focused on estrogen receptor mediated pathways and did not comprehensively clarify the observed toxicity. O-GlcNAcase (OGA), the highest level in brain, plays a critical role in controlling neuronal functions at multi-levels from molecule to animal behaviors. In this work, we intend to investigate the underlying molecular mechanisms for the neurotoxicity of bisphenol analogues by identifying their cellular targets and the resultant effects. The inhibitory actions of seven bisphenol analogues on the OGA activity at molecular level were investigated by our developed electrochemical biosensor. We found that their potency varied with substituent groups, in which tetrabromo bisphenol A (TBBPA) was the strongest. The seven bisphenol analogues (0-100 µM exposure) significantly inhibited OGA activity and up-regulated protein O-GlcNAcylation level in PC12 cells. Inhibition of OGA by bisphenol analogues further induced intracellular calcium, ROS, inflammation, repressed proliferation, interfered with cell cycle, induced apoptosis. And especially, 10 µM tetrabromo bisphenol A (TBBPA) exposure could impair the growth and development of neurite in human neural stem cells (hNSCs). Molecular docking for OGA/bisphenol analogue complexes revealed the hydrophobicity-dominated inhibition potency. OGA, as a new cellular target of bisphenol analogues, would illuminate the molecular mechanism of bisphenol analogues neurotoxicity.


Benzhydryl Compounds/toxicity , Environmental Pollutants/toxicity , Neural Stem Cells/drug effects , Neurotoxicity Syndromes/enzymology , Phenols/toxicity , beta-N-Acetylhexosaminidases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Benzhydryl Compounds/chemistry , Calcium/metabolism , Cell Proliferation/drug effects , Cytokines/metabolism , Dose-Response Relationship, Drug , Environmental Pollutants/chemistry , Humans , Molecular Docking Simulation , Neural Stem Cells/enzymology , Neural Stem Cells/immunology , Neuronal Outgrowth/drug effects , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/immunology , PC12 Cells , Phenols/chemistry , Rats , Reactive Oxygen Species/metabolism
18.
Neurotoxicology ; 74: 81-90, 2019 09.
Article En | MEDLINE | ID: mdl-31175943

BACKGROUND: Sixty percent of the mammalian brain is composed of lipids including arachidonic acid (AA). AA released from cell membranes is metabolised in the cyclooxygenase (COX) pathway to prostanoids - biologically active substances involved in the regulation of many processes including inflammation. It has been shown that long-term exposure to fluoride in pre and neonatal period is dangerous because this element is able to penetrate through the placenta and to cross the blood-brain barrier. Exposure to fluoride during the development affects metabolism and physiology of neurons and glia which results in the impairment of cognitive functions but the exact mechanisms of fluoride neurotoxicity are not clearly defined. OBJECTIVE: The aim of this study was to determine whether exposure to fluoride during the development affects COXes activity and the synthesis of prostanoids. MATERIAL AND METHODS: Pre- and postnatal toxicity model in Wistar rats was used. Experimental animals received 50 mg/L of NaF in drinking water ad libitum, while control animals received tap water. In cerebral cortex, hippocampus, cerebellum and striatum were measured fluoride concentration, COX1 and COX2 genes expression, immunolocalization of the enzymatic proteins and concentration of PGE2 and TXB2. RESULTS: of this study showed statistically significant changes in the concentration of fluoride in brain structures between study group and control animals. Moreover, significant changes in the expression level of COX1 and COX2, and in the concentration of PGE2 and TXB2 were observed. CONCLUSION: Exposure to fluoride in the prenatal and neonatal period result in the increase in COX2 activity and increase in PGE2 concentration in rats brain, which may lead to disturbances in central nervous system homeostasis.‬‬.


Cyclooxygenase 1/biosynthesis , Cyclooxygenase 1/genetics , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Fluorides/toxicity , Gene Expression Regulation, Enzymologic/drug effects , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/genetics , Animals , Animals, Newborn , Brain/drug effects , Brain/enzymology , Brain/metabolism , Dinoprostone/biosynthesis , Female , Fluorides/pharmacokinetics , Pregnancy , Prenatal Exposure Delayed Effects/enzymology , Prenatal Exposure Delayed Effects/genetics , Prostaglandins/biosynthesis , Rats , Rats, Wistar , Thromboxane B2/biosynthesis
19.
Curr Drug Saf ; 14(3): 209-216, 2019.
Article En | MEDLINE | ID: mdl-31057112

BACKGROUND: Cyclophosphamide (CPA) is the most widely prescribed cancer chemotherapeutic agent which shows serious neurotoxic side effect. Generation of reactive oxygen species at the cellular level is the basic mechanism of cyclophosphamide induced neurotoxicity. Edaravone is the synthetic drug used for brain stroke and has potent antioxidant property. OBJECTIVE: This study aimed to investigate the effect of edaravone on neurobehavioral and neuropathological alteration induced by cyclophosphamide in male rats. METHODS: Twenty eight Sprague-Dawley rats were equally divided into four groups of seven rats in each. The control group received saline, and other groups were given CPA intraperitoneally (100 mg/kg), CPA (100 mg/kg) intraperitoneally + Edaravone (10 mg/kg) orally, or Edaravone (10 mg/kg) orally for one month. RESULTS: Our data showed that CPA significantly elevated brain AChE activity in the hippocampal region. A decrease in the total antioxidant capacity and a reduction in the CAT, SOD, and GPX activity occurred in the brains of the rats exposed to CPA. CPA-treated rats showed a significant impairment in long-termmemory and motor coordination. These results were supported by histopathological observations of the brain. Results revealed that administration of edaravone reversed AChE activity alternation and ameliorated behavioral and histopathological changes induced by CPA. CONCLUSION: This study suggests that co-administration of edaravone with cyclophosphamide may be a useful intriguing therapeutic approach to overcome cyclophosphamide induced neurotoxicity.


Cyclophosphamide/toxicity , Edaravone/pharmacology , Neurotoxicity Syndromes/prevention & control , Oxidative Stress/drug effects , Acetylcholinesterase/metabolism , Animals , GPI-Linked Proteins/metabolism , Male , Neurotoxicity Syndromes/enzymology , Rats, Sprague-Dawley
20.
Chemosphere ; 226: 817-824, 2019 Jul.
Article En | MEDLINE | ID: mdl-30965253

Reptiles, the most diverse taxon of terrestrial vertebrates, might be particularly vulnerable to soil pollution. Reptiles especially lizards have been rarely evaluated in ecotoxicological studies, and there is a very limited report for effects of soil pesticide contaminants on lizards. In this study, male and female lizards (Eremias argus) were exposed to Glufosinate-ammonium (GLA) and l- Glufosinate-ammonium (L-GLA) for 60 days. Slower sprint speed, higher frequency of turning back and reduced brain index were observed in treatment groups. The accumulation of GLA in the brain of lizard was higher than that of L-GLA. Moreover, the activities of neurotoxicity-related enzymes and biomarkers of oxidative stress were also investigated. In summary, the neurotoxic effects of lizards have been observed after exposure to GLA and L-GLA. Based on the result of the Integrated Biomarker Response (IBR), males were more sensitive to contaminants than females. On the other hand, the neurotoxic pathways by GLA and L-GLA triggered were slightly different: GLA mainly acted on glutamine synthetase (GS), acetylcholinesterase (AchE) and Catalase (CAT) and L-GLA aimed at AchE, Na+/K+-ATPase, Superoxide dismutase (SOD) and Malondialdehyde (MDA). In summary, the accumulation of GLA and L-GLA in lizard's brain induced neurotoxicity by altering the levels of enzymes related to nervous system and antioxidant activity and further resulted in the decrease of brain index and locomotor performance.


Aminobutyrates/toxicity , Environmental Pollution/adverse effects , Lizards/metabolism , Locomotion/drug effects , Neurotoxicity Syndromes/etiology , Aminobutyrates/pharmacokinetics , Aminobutyrates/pharmacology , Animals , Brain/enzymology , Brain/metabolism , Ecotoxicology , Female , Lizards/physiology , Male , Neurotoxicity Syndromes/enzymology , Oxidative Stress/drug effects , Pesticides/metabolism , Sex Factors
...