Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.241
Filtrer
1.
Int J Mol Sci ; 25(11)2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38891838

RÉSUMÉ

Nanoparticles (NPs) are becoming increasingly important novel materials for many purposes, including basic research, medicine, agriculture, and engineering. Increasing human and environmental exposure to these promising compounds requires assessment of their potential health risks. While the general direct cytotoxicity of NPs is often routinely measured, more indirect possible long-term effects, such as reproductive or developmental neurotoxicity (DNT), have been studied only occasionally and, if so, mostly on non-human animal models, such as zebrafish embryos. In this present study, we employed a well-characterized human neuronal precursor cell line to test the concentration-dependent DNT of green-manufactured copper sulfide (CuS) nanoparticles on crucial early events in human brain development. CuS NPs turned out to be generally cytotoxic in the low ppm range. Using an established prediction model, we found a clear DNT potential of CuS NPs on neuronal precursor cell migration and neurite outgrowth, with IC50 values 10 times and 5 times, respectively, lower for the specific DNT endpoint than for general cytotoxicity. We conclude that, in addition to the opportunities of NPs, their risks to human health should be carefully considered.


Sujet(s)
Cuivre , Nanoparticules métalliques , Neurones , Humains , Cuivre/toxicité , Nanoparticules métalliques/toxicité , Nanoparticules métalliques/composition chimique , Neurones/effets des médicaments et des substances chimiques , Sulfures/toxicité , Sulfures/composition chimique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Syndromes neurotoxiques/étiologie , Syndromes neurotoxiques/anatomopathologie , Nanoparticules/toxicité , Nanoparticules/composition chimique , Cellules souches neurales/effets des médicaments et des substances chimiques , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques
2.
Crit Rev Toxicol ; 54(5): 330-343, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38832580

RÉSUMÉ

Despite the growing epidemiological evidence of an association between toxin exposure and developmental neurotoxicity (DNT), systematic testing of DNT is not mandatory in international regulations for admission of pharmaceuticals or industrial chemicals. However, to date around 200 compounds, ranging from pesticides, pharmaceuticals and industrial chemicals, have been tested for DNT in the current OECD test guidelines (TG-443 or TG-426). There are calls for the development of new approach methodologies (NAMs) for DNT, which has resulted in a DNT testing battery using in vitro human cell-based assays. These assays provide a means to elucidate the molecular mechanisms of toxicity in humans which is lacking in animal-based toxicity tests. However, cell-based assays do not represent all steps of the complex process leading to DNT. Validated models with a multi-organ network of pathways that interact at the molecular, cellular and tissue level at very specific timepoints in a life cycle are currently missing. Consequently, whole model organisms are being developed to screen for, and causally link, new molecular targets of DNT compounds and how they affect whole brain development and neurobehavioral endpoints. Given the practical and ethical restraints associated with vertebrate testing, lower animal models that qualify as 3 R (reduce, refine and replace) models, including the nematode (Caenorhabditis elegans) and the zebrafish (Danio rerio) will prove particularly valuable for unravelling toxicity pathways leading to DNT. Although not as complex as the human brain, these 3 R-models develop a complete functioning brain with numerous neurodevelopmental processes overlapping with human brain development. Importantly, the main signalling pathways relating to (neuro)development, metabolism and growth are highly conserved in these models. We propose the use of whole model organisms specifically zebrafish and C. elegans for DNT relevant endpoints.


Sujet(s)
Caenorhabditis elegans , Syndromes neurotoxiques , Tests de toxicité , Danio zébré , Animaux , Caenorhabditis elegans/effets des médicaments et des substances chimiques , Modèles animaux , Tests de toxicité/méthodes
3.
Turk Psikiyatri Derg ; 35(2): 150-155, 2024.
Article de Anglais, Turc | MEDLINE | ID: mdl-38842156

RÉSUMÉ

Lithium may cause toxicity as it has a narrow therapeutic range. Lithium intoxication may manifest in the form of acute, acute on chronic and chronic intoxication. Neurotoxicity is a common component of chronic lithium intoxication and the symptoms include tremor, ataxia, dysarthria, extrapyramidal symptoms, hyperreflexia, seizures and status epilepticus. Although rare, catatonia could as a manifestation of lithium neurotoxicity. In this report, we present a patient with bipolar disorder presenting with catatonic symptoms secondary to lithium intoxication. We will discuss the risk factors, differential diagnosis and the treatment of catatonic symptoms. Lithium neurotoxicity may present with various clinical symptoms including catatonia, and differential diagnosis should be made well in such cases. If lithium neurotoxicity is suspected, rapid and appropriate intervention is required to prevent permanent neurological damage. Keywords: Lithium, Neurotoxicity, Catatonia.


Sujet(s)
Trouble bipolaire , Catatonie , Humains , Antimaniacodépressifs/effets indésirables , Trouble bipolaire/traitement médicamenteux , Catatonie/induit chimiquement , Diagnostic différentiel , Syndromes neurotoxiques/étiologie , Syndromes neurotoxiques/diagnostic
4.
Int J Mol Sci ; 25(12)2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38928257

RÉSUMÉ

The peripheral nervous system can encounter alterations due to exposure to some of the most commonly used anticancer drugs (platinum drugs, taxanes, vinca alkaloids, proteasome inhibitors, thalidomide), the so-called chemotherapy-induced peripheral neurotoxicity (CIPN). CIPN can be long-lasting or even permanent, and it is detrimental for the quality of life of cancer survivors, being associated with persistent disturbances such as sensory loss and neuropathic pain at limb extremities due to a mostly sensory axonal polyneuropathy/neuronopathy. In the state of the art, there is no efficacious preventive/curative treatment for this condition. Among the reasons for this unmet clinical and scientific need, there is an uncomplete knowledge of the pathogenetic mechanisms. Ion channels and transporters are pivotal elements in both the central and peripheral nervous system, and there is a growing body of literature suggesting that they might play a role in CIPN development. In this review, we first describe the biophysical properties of these targets and then report existing data for the involvement of ion channels and transporters in CIPN, thus paving the way for new approaches/druggable targets to cure and/or prevent CIPN.


Sujet(s)
Antinéoplasiques , Canaux ioniques , Neuropathies périphériques , Humains , Antinéoplasiques/effets indésirables , Neuropathies périphériques/induit chimiquement , Neuropathies périphériques/métabolisme , Canaux ioniques/métabolisme , Animaux , Syndromes neurotoxiques/étiologie , Syndromes neurotoxiques/métabolisme , Protéines de transport membranaire/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme
5.
Int Immunopharmacol ; 136: 112335, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-38815349

RÉSUMÉ

Chlorpyrifos (CPF) is a widely used organophosphate insecticide in agriculture and homes. Exposure to organophosphates is associated with neurotoxicity. Fluoxetine (FLX) is a selective serotonin reuptake inhibitor (SSRI) that is widely prescribed for depression and anxiety disorders. Studies have shown that FLX has neuroprotective, anti-inflammatory, antioxidant, and antiapoptotic effects. The molecular mechanisms underlying FLX are not fully understood. This work aimed to investigate the potential neuroprotective effect of FLX on CPF-induced neurotoxicity and the underlying molecular mechanisms involved. Thirty-two rats were randomly divided into four groups: (I) the vehicle control group; (II) the FLX-treated group (10 mg/kg/day for 28 days, p.o); (III) the CPF-treated group (10 mg/kg for 28 days); and (IV) the FLX+CPF group. FLX attenuated CPF-induced neuronal injury, as evidenced by a significant decrease in Aß and p-Tau levels and attenuation of cerebral and hippocampal histological abrasion injury induced by CPF. FLX ameliorated neuronal oxidative stress, effectively reduced MDA production, and restored SOD and GSH levels through the coactivation of the PPARγ and SIRT1 proteins. FLX counteracted the neuronal inflammation induced by CPF by decreasing MPO, NO, TNF-α, IL-1ß, and IL-6 levels by suppressing NF-κB and JAK1/STAT3 activation. The antioxidant and anti-inflammatory properties of FLX help to prevent CPF-induced neuronal intoxication.


Sujet(s)
Chlorpyriphos , Fluoxétine , Janus kinase 1 , Facteur de transcription NF-kappa B , Neuroprotecteurs , Récepteur PPAR gamma , Facteur de transcription STAT-3 , Transduction du signal , Sirtuine-1 , Animaux , Facteur de transcription STAT-3/métabolisme , Sirtuine-1/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Récepteur PPAR gamma/métabolisme , Janus kinase 1/métabolisme , Mâle , Fluoxétine/pharmacologie , Fluoxétine/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques , Chlorpyriphos/toxicité , Rats , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Neurones/effets des médicaments et des substances chimiques , Neurones/anatomopathologie , Stress oxydatif/effets des médicaments et des substances chimiques , Insecticides/toxicité , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Rat Sprague-Dawley , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Hippocampe/anatomopathologie , Syndromes neurotoxiques/traitement médicamenteux , Syndromes neurotoxiques/anatomopathologie
6.
Chemosphere ; 359: 142246, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38710414

RÉSUMÉ

The knowledge and assessment of mixtures of chemical pollutants in the aquatic environment is a complex issue that is often challenging to address. In this review, we focused on the use of zebrafish (Danio rerio), a vertebrate widely used in biomedical research, as a model for detecting the effects of chemical mixtures with a focus on behaviour. Our aim was to summarize the current status of the ecotoxicological research in this sector. Specifically, we limited our research to the period between January 2012 and September 2023, including only those works aimed at detecting neurotoxicity through behavioural endpoints, utilizing zebrafish at one or more developmental stages, from egg to adult. Additionally, we gathered the findings for every group of chemicals involved and summarised data from all the works we included. At the end of the screening process 101 papers were considered eligible for inclusion. Results show a growing interest in zebrafish at all life stages for this kind of research in the last decade. Also, a wide variety of different assays, involving different senses, was used in the works we surveyed, with exposures ranging from acute to chronic. In conclusion, the results of this study show the versatility of zebrafish as a model for the detection of mixture toxicity although, for what concerns behavioural analysis, the lack of standardisation of methods and endpoints might still be limiting.


Sujet(s)
Comportement animal , Syndromes neurotoxiques , Polluants chimiques de l'eau , Danio zébré , Animaux , Polluants chimiques de l'eau/toxicité , Comportement animal/effets des médicaments et des substances chimiques , Syndromes neurotoxiques/étiologie , Tests de toxicité/méthodes , Écotoxicologie/méthodes
7.
Basic Clin Pharmacol Toxicol ; 135(1): 81-97, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38780039

RÉSUMÉ

We established experimental models of manganese (Mn) and iron (Fe) exposure in vitro and in vivo, and addressed the effects of manganese and iron combined exposure on the synaptic function of pheochromocytoma derived cell line 12 (PC12) cells and rat cortex, respectively. We investigated the protective effect of sodium para-aminosalicylate (PAS-Na) on manganese and iron combined neurotoxicity, providing a scientific basis for the prevention and treatment of ferromanganese combined neurotoxicity. Western blot and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were performed to detect the expression levels of protein and mRNA related to synaptic damage. Y-maze novelty test and balance beam test were used to evaluate the motor and cognitive function of rats. Haematoxylin and eosin (H&E) and Nissl staining were performed to observe the cortical damage of rats. The results showed that the combined exposure of Mn and Fe in rats led to a synergistic effect, attenuating growth and development, and altering learning and memory as well as motor function. The combination of Mn and Fe also caused damage to the synaptic structure of PC12 cells, which is manifested as swelling of dendrites and axon terminals, and even lead to cell death. PAS-Na displayed some antagonistic effects against the Mn- and Fe-induced synaptic structural damage, growth, learning and memory impairment.


Sujet(s)
Acide aminosalicylique , Manganèse , Synapses , Animaux , Rats , Cellules PC12 , Synapses/effets des médicaments et des substances chimiques , Mâle , Acide aminosalicylique/pharmacologie , Manganèse/toxicité , Cortex cérébral/effets des médicaments et des substances chimiques , Cortex cérébral/anatomopathologie , Cortex cérébral/métabolisme , Rat Sprague-Dawley , Fer/métabolisme , Neuroprotecteurs/pharmacologie , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques , Syndromes neurotoxiques/prévention et contrôle , Syndromes neurotoxiques/étiologie , Syndromes neurotoxiques/anatomopathologie , Modèles animaux de maladie humaine
8.
Environ Res ; 256: 119237, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38810829

RÉSUMÉ

Ionizing radiation (IR) poses a significant threat to both the natural environment and biological health. Exposure to specific doses of ionizing radiation early in an organism's development can lead to developmental toxicity, particularly neurotoxicity. Through experimentation with Xenopus laevis (X. laevis), we examined the effects of radiation on early developmental stage. Our findings revealed that radiation led to developmental abnormalities and mortality in X. laevis embryos in a dose-dependent manner, disrupting redox homeostasis and inducing cell apoptosis. Additionally, radiation caused neurotoxic effects, resulting in abnormal behavior and neuron damage in the embryos. Further investigation into the underlying mechanisms of radiation-induced neurotoxicity indicated the potential involvement of the neuroactive ligand-receptor interaction pathway, which was supported by RNA-Seq analysis. Validation of gene expression associated with this pathway and analysis of neurotransmitter levels confirmed our hypothesis. In addition, we further validated the important role of this signaling pathway in radiation-induced neurotoxicity through edaravone rescue experiments. This research establishes a valuable model for radiation damage studying and provides some insight into radiation-induced neurotoxicity mechanisms.


Sujet(s)
Embryon non mammalien , Rayonnement ionisant , Xenopus laevis , Animaux , Embryon non mammalien/effets des radiations , Syndromes neurotoxiques/étiologie , Transduction du signal/effets des radiations , Apoptose/effets des radiations , Ligands
9.
Environ Pollut ; 355: 124280, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38815890

RÉSUMÉ

Cr(VI) is a common hazardous heavy metal contaminant that seriously endangers human and aquatic animal health. GPX4 was the key enzyme that reduces heavy metal toxicity through inhibiting ferroptosis pathway. Astaxanthin was GPX4 activator that can weaken biological toxicity induced by Cr(VI) exposure. The present study was conducted to evaluate the major role of GPX4 in astaxanthin protects Cr(VI)-induced oxidative damage, blood-brain barrier injury and neurotoxicity in brain-liver axis through inhibiting ferroptosis pathway. In the current study, astaxanthin intervention can effectively alleviate Cr(VI)-induced oxidative stress, blood-brain barrier damage, and neurotoxicity. GPX4 plays a major role in mediating astaxanthin nutritional intervention to reduce ROS and liver non-heme iron accumulation, which would contribute to the reduction of ferroptosis. Meanwhile, astaxanthin maintains the stability of transport receptors and protein macromolecules such as TMEM163, SLC7A11, SLC3A2, FPN1 and GLUT1 in the brain liver axis, promoting substance exchange and energy supply. Moreover, astaxanthin alleviates Cr(VI)-induced neurotoxicity by promoting tight protein expression and reducing blood-brain barrier permeability.


Sujet(s)
Barrière hémato-encéphalique , Chrome , Polluants chimiques de l'eau , Xanthophylles , Danio zébré , Xanthophylles/pharmacologie , Animaux , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Chrome/toxicité , Polluants chimiques de l'eau/toxicité , Stress oxydatif/effets des médicaments et des substances chimiques , Syndromes neurotoxiques/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme
10.
J Hazard Mater ; 473: 134607, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38761765

RÉSUMÉ

Paraquat (PQ) exposure is strongly associated with neurotoxicity. However, research on the neurotoxicity mechanisms of PQ varies in terms of endpoints of toxic assessment, resulting in a great challenge to understand the early neurotoxic effects of PQ. In this study, we developed an adverse outcome pathway (AOP) to investigate PQ-induced neuro-immunotoxicity from an immunological perspective, combining of traditional toxicology methods and computer simulations. In vivo, PQ can microstructurally lead to an early synaptic loss in the brain mice, which is a large degree regarded as a main reason for cognitive impairment to mice behavior. Both in vitro and in vivo demonstrated synapse loss is caused by excessive activation of the complement C1q/C3-CD11b pathway, which mediates microglial phagocytosis dysfunction. Additionally, the interaction between PQ and C1q was validated by molecular simulation docking. Our findings extend the AOP framework related to PQ neurotoxicity from a neuro-immunotoxic perspective, highlighting C1q activation as the initiating event for PQ-induced neuro-immunotoxicity. In addition, downstream complement cascades induce abnormal microglial phagocytosis, resulting in reduced synaptic density and subsequent non-motor dysfunction. These findings deepen our understanding of neurotoxicity and provide a theoretical basis for ecological risk assessment of PQ.


Sujet(s)
Complément C1q , Simulation numérique , Microglie , Paraquat , Phagocytose , Paraquat/toxicité , Animaux , Complément C1q/immunologie , Complément C1q/métabolisme , Phagocytose/effets des médicaments et des substances chimiques , Microglie/effets des médicaments et des substances chimiques , Voies des issues indésirables , Mâle , Syndromes neurotoxiques/immunologie , Syndromes neurotoxiques/anatomopathologie , Syndromes neurotoxiques/étiologie , Souris , Encéphale/effets des médicaments et des substances chimiques , Herbicides/toxicité , Antigènes CD11b/métabolisme , Complément C3/métabolisme , Simulation de docking moléculaire , Synapses/effets des médicaments et des substances chimiques , Souris de lignée C57BL
11.
Zool Res ; 45(3): 691-703, 2024 May 18.
Article de Anglais | MEDLINE | ID: mdl-38766750

RÉSUMÉ

General anesthetic agents can impact brain function through interactions with neurons and their effects on glial cells. Oligodendrocytes perform essential roles in the central nervous system, including myelin sheath formation, axonal metabolism, and neuroplasticity regulation. They are particularly vulnerable to the effects of general anesthetic agents resulting in impaired proliferation, differentiation, and apoptosis. Neurologists are increasingly interested in the effects of general anesthetic agents on oligodendrocytes. These agents not only act on the surface receptors of oligodendrocytes to elicit neuroinflammation through modulation of signaling pathways, but also disrupt metabolic processes and alter the expression of genes involved in oligodendrocyte development and function. In this review, we summarize the effects of general anesthetic agents on oligodendrocytes. We anticipate that future research will continue to explore these effects and develop strategies to decrease the incidence of adverse reactions associated with the use of general anesthetic agents.


Sujet(s)
Anesthésiques généraux , Encéphale , Oligodendroglie , Oligodendroglie/effets des médicaments et des substances chimiques , Animaux , Encéphale/effets des médicaments et des substances chimiques , Anesthésiques généraux/effets indésirables , Anesthésiques généraux/toxicité , Syndromes neurotoxiques/étiologie , Humains
12.
Nat Rev Clin Oncol ; 21(7): 501-521, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38769449

RÉSUMÉ

Chimeric antigen receptor (CAR) T cell therapy has revolutionized the treatment of several haematological malignancies and is being investigated in patients with various solid tumours. Characteristic CAR T cell-associated toxicities such as cytokine-release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) are now well-recognized, and improved supportive care and management with immunosuppressive agents has made CAR T cell therapy safer and more feasible than it was when the first regulatory approvals of such treatments were granted in 2017. The increasing clinical experience with these therapies has also improved recognition of previously less well-defined toxicities, including movement disorders, immune effector cell-associated haematotoxicity (ICAHT) and immune effector cell-associated haemophagocytic lymphohistiocytosis-like syndrome (IEC-HS), as well as the substantial risk of infection in patients with persistent CAR T cell-induced B cell aplasia and hypogammaglobulinaemia. A more diverse selection of immunosuppressive and supportive-care pharmacotherapies is now being utilized for toxicity management, yet no universal algorithm for their application exists. As CAR T cell products targeting new antigens are developed, additional toxicities involving damage to non-malignant tissues expressing the target antigen are a potential hurdle. Continued prospective evaluation of toxicity management strategies and the design of less-toxic CAR T cell products are both crucial for ongoing success in this field. In this Review, we discuss the evolving understanding and clinical management of CAR T cell-associated toxicities.


Sujet(s)
Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Humains , Immunothérapie adoptive/effets indésirables , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/usage thérapeutique , Syndrome de libération de cytokines/étiologie , Syndrome de libération de cytokines/immunologie , Syndromes neurotoxiques/étiologie , Syndromes neurotoxiques/immunologie , Tumeurs hématologiques/thérapie , Tumeurs hématologiques/immunologie , Tumeurs/immunologie , Tumeurs/thérapie , Lymphocytes T/immunologie
13.
Clin Neurophysiol ; 163: 132-142, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38733703

RÉSUMÉ

BACKGROUND: Immune effector cell-associated neurotoxicity syndrome (ICANS) is common after chimeric antigen receptor T-cell (CAR-T) therapy. OBJECTIVE: This study aimed to assess the impact of preinfusion electroencephalography (EEG) abnormalities and EEG findings at ICANS onset for predicting ICANS risk and severity in 56 adult patients with refractory lymphoma undergoing CAR-T therapy. STUDY DESIGN: EEGs were conducted at the time of lymphodepleting chemotherapy and shortly after onset of ICANS. RESULTS: Twenty-eight (50%) patients developed ICANS at a median time of 6 days after CAR-T infusion. Abnormal preinfusion EEG was identified as a risk factor for severe ICANS (50% vs. 17%, P = 0.036). Following ICANS onset, EEG abnormalities were detected in 89% of patients [encephalopathy (n = 19, 70%) and/or interictal epileptiform discharges (IEDs) (n = 14, 52%)]. Importantly, IEDs seemed to be associated with rapid progression to higher grades of ICANS within 24 h. CONCLUSIONS: If confirmed in a large cohort of patients, these findings could establish the basis for modifying current management guidelines, enabling the identification of patients at risk of neurotoxicity, and providing support for preemptive corticosteroid use in patients with both initial grade 1 ICANS and IEDs at neurotoxicity onset, who are at risk of neurological impairment.


Sujet(s)
Électroencéphalographie , Immunothérapie adoptive , Syndromes neurotoxiques , Humains , Mâle , Femelle , Adulte d'âge moyen , Syndromes neurotoxiques/physiopathologie , Syndromes neurotoxiques/étiologie , Syndromes neurotoxiques/diagnostic , Adulte , Immunothérapie adoptive/effets indésirables , Immunothérapie adoptive/méthodes , Sujet âgé , Lymphomes/thérapie , Lymphomes/physiopathologie , Lymphomes/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Jeune adulte
14.
Food Chem Toxicol ; 189: 114747, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38768937

RÉSUMÉ

Chronic exposure to lead (Pb) induces neurodegenerative changes in animals and humans. Drugs with strong antioxidant properties are effective against Pb-mediated neurotoxicity. In a prior study, we identified 5,7-dihydroxy-3',4',5'-trimethoxyflavone (TMF) from Ocimum basilicum L. leaves as a potent antioxidant and neuroprotective compound. This research explores TMF's neuroprotective effects against Pb-induced brain toxicity in rats to establish it as a therapeutic agent. Rats received lead acetate (100 mg/kg, orally, once daily) for 30 days to induce brain injury, followed by TMF treatment (5 and 10 mg/kg, oral, once daily) 30 min later. Cognitive and motor functions were assessed using Morris Water Maze and horizontal bar tests. Lead, monoamine oxidase (MAO) A and B enzymes, reduced glutathione (GSH), thiobarbituric acid reactive species (TBARS), Tumor necrosis factor-alpha (TNF-α), and IL-6 levels were measured in the hippocampus and cerebellum. Pb exposure impaired cognitive and motor functions, increased Pb, TBARS, TNF-α, and IL-6 levels, and compromised MAO A & B and GSH levels. TMF reversed Pb-induced memory and motor deficits and normalized biochemical anomalies. TMF's neuroprotective effects against lead involve chelating, antioxidant, anti-inflammatory, and monoaminergic properties, suggesting its potential as a treatment for metal-induced brain injury.


Sujet(s)
Anti-inflammatoires , Antioxydants , Animaux , Antioxydants/pharmacologie , Mâle , Rats , Anti-inflammatoires/pharmacologie , Neuroprotecteurs/pharmacologie , Flavones/pharmacologie , Plomb/toxicité , Chélateurs/pharmacologie , Rat Wistar , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Syndromes neurotoxiques/traitement médicamenteux , Syndromes neurotoxiques/prévention et contrôle , Glutathion/métabolisme , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques
15.
Brain Res ; 1839: 149017, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-38768935

RÉSUMÉ

Parkinson's disease (PD) is a complex disorder, primarily of idiopathic origin, with environmental stressors like rotenone and manganese linked to its development. This study explores their potential interaction and resulting neurotoxicity, aiming to understand how environmental factors contribute to PD. In an eight-day experiment, male Wistar rats weighing 280-300 g were subjected to rotenone, manganese, or a combination of both. Various parameters were assessed, including body weight, behavior, serum markers, tissue damage, protein levels (tyrosine hydroxylase, Dopamine- and cAMP-regulated neuronal phosphoprotein -DARPP-32-, and α-synuclein), and mitochondrial function. Manganese heightened rotenone's impact on reducing food intake without causing kidney or liver dysfunction. However, the combined exposure intensified neurotoxicity, which was evident in augmented broken nuclei and decreased tyrosine hydroxylase and DARPP-32 levels in the striatum. While overall mitochondrial function was preserved, co-administration reduced complex IV activity in the midbrain and liver. In conclusion, our findings revealed a parallel toxic effect induced by rotenone and manganese. Notably, while these substances do not target the same dopaminergic regions, a notable escalation in toxicity is evident in the striatum, the brain region where their toxic effects converge. This study highlights the need for further exploration regarding the interaction of environmental factors and their possible impact on the etiology of PD.


Sujet(s)
Manganèse , Rat Wistar , Roténone , Tyrosine 3-monooxygenase , Animaux , Roténone/toxicité , Mâle , Manganèse/toxicité , Rats , Tyrosine 3-monooxygenase/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Phosphoprotéine DARPP-32 régulée par la dopamine et l'AMPc/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , alpha-Synucléine/métabolisme , Syndromes neurotoxiques/métabolisme , Corps strié/métabolisme , Corps strié/effets des médicaments et des substances chimiques
16.
Int Immunopharmacol ; 135: 112336, 2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-38801809

RÉSUMÉ

IIrbesartan (IRB), an angiotensin II type 1 receptor (AT1R) antagonist, has been widely employed in the medical field for its effectiveness in managing hypertension. However, there have been no documented investigations regarding the immunostimulatory properties of IRB. To address this gap, this study has been performed to assess the neuroprotective impact of IRB as an immunostimulatory agent in mitigating acute neurotoxicity induced by cyclophosphamide (CYP) in rats. mRNA levels of nuclear factor erythroid 2 (Nrf-2), interleukin (IL)-18, IL-1ß, and MMP-1 have been assessed using quantitative real-time polymerase chain reaction (qRT-PCR). Additionally, the levels of malondialdehyde (MDA), reduced glutathione (GSH), and superoxide dismutase (SOD) has been evaluated to assess the oxidative stress. Additionally, macrophage inflammatory protein 2 (MIP2) has been evaluated using enzyme-linked immunosorbent assay (ELISA). Western blotting has been used to investigate the protein expression of nucleotide binding oligomerization domain-like receptor protein 3 (NLRP3) and caspase-1 (CASP-1), along with an assessment of histopathological changes. Administration of IRB protected against oxidative stress by augmenting the levels of GSH and SOD as well as reducing MDA level. Also, administration of IRB led to a diminishment in the brain levels of MIP2 and MMP1. Furthermore, it led to a suppression of IL-1ß and IL-18 levels, which are correlated with a reduction in the abundance of NLRP3 and subsequently CASP-1. This study provides new insights into the immunomodulatory effects of IRB in the context of CYP-induced acute neurotoxicity. Specifically, IRB exerts its effects by reducing oxidative stress, neuroinflammation, inhibiting chemokine recruitment, and mitigating neuronal degeneration through the modulation of immune markers. Therefore, it can be inferred that the use of IRB as an immunomodulator has the potential to effectively mitigate immune disorders associated with inflammation.


Sujet(s)
Cyclophosphamide , Inflammasomes , Irbésartan , Protéine-3 de la famille des NLR contenant un domaine pyrine , Stress oxydatif , Animaux , Cyclophosphamide/toxicité , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Inflammasomes/métabolisme , Inflammasomes/effets des médicaments et des substances chimiques , Irbésartan/pharmacologie , Irbésartan/usage thérapeutique , Mâle , Rats , Stress oxydatif/effets des médicaments et des substances chimiques , Syndromes neurotoxiques/traitement médicamenteux , Syndromes neurotoxiques/immunologie , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques , Immunomodulation/effets des médicaments et des substances chimiques , Rat Wistar
17.
Sci Total Environ ; 935: 173457, 2024 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-38782285

RÉSUMÉ

Microplastics and chlorine-containing triclosan (TCS) are widespread in aquatic environments and may pose health risks to organisms. However, studies on the combined toxicity of aged microplastics and TCS are limited. To investigate the toxic effects and potential mechanisms associated with co-exposure to TCS adsorbed on aged polyethylene microplastics (aPE-MPs) at environmentally relevant concentrations, a 7-day chronic exposure experiment was conducted using Xenopus tropicalis tadpoles. The results showed that the overall particle size of aPE-MPs decreased after 30 days of UV aging, whereas the increase in specific surface area improved the adsorption capacity of aPE-MPs for TCS, resulting in the bioaccumulation of TCS under dual-exposure conditions in the order of aPE-TCS > PE-TCS > TCS. Co-exposure to aPE-MPs and TCS exacerbated oxidative stress and neurotoxicity to a greater extent than a single exposure. Significant upregulation of pro-symptomatic factors (IL-ß and IL-6) and antioxidant enzyme activities (SOD and CAT) indicated that the aPE-TCS combination caused more severe oxidative stress and inflammation. Molecular docking revealed the molecular mechanism of the direct interaction between TCS and SOD, CAT, and AChE proteins, which explains why aPE-MPs promote the bioaccumulation of TCS, causing increased toxicity upon combined exposure. These results emphasize the need to be aware of the combined toxicity caused by the increased ability of aged microplastics to carry contaminants.


Sujet(s)
Larve , Microplastiques , Stress oxydatif , Triclosan , Polluants chimiques de l'eau , Xenopus , Animaux , Microplastiques/toxicité , Polluants chimiques de l'eau/toxicité , Triclosan/toxicité , Larve/effets des médicaments et des substances chimiques , Bioaccumulation , Syndromes neurotoxiques
18.
Int Immunopharmacol ; 135: 112308, 2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-38788447

RÉSUMÉ

Although colistin has a crucial antibacterial activity in treating multidrug-resistant gram-negative bacteria strains; it exhibited renal and neuronal toxicities rendering its use a challenge. Previous studies investigated the incretin hormones either glucose-dependent insulinotropic polypeptide (GIP) or glucagonlike peptide-1 (GLP-1) for their neuroprotective and nephroprotective effectiveness. The present study focused on investigating Tirzepatide (Tirze), a dual GLP-1/GIP agonist, as an adjuvant therapy in the colistin treatment protocol for attenuating its renal and neuronal complications. Rats were divided into; The normal control group, the colistin-treated group received colistin (300,000 IU/kg/day for 7 days; i.p.). The Tirze-treated group received Tirze (1.35 mg/kg on the 1,4,7thdays; s.c.) and daily colistin. Tirze effectively enhanced histopathological alterations, renal function parameters, and locomotor activity in rats. Tirze mechanistically acted via modulating various signaling axes evolved under the insult of phosphatidylinositol 3-kinases (PI3K)/phosphorylated protein kinase-B (p-Akt)/ glycogen synthase kinase (GSK)3-ß hub causing mitigation of nuclear factor (NF)-κB (NF-κB) / tumor necrosis factor-α (TNF-α), increment of nuclear factor erythroid 2-related factor 2 (Nrf2)/ glutathione (GSH), downregulation of ER stress-related biomarkers (activation transcription factor 4 (ATF4) and C/EBP homologous protein (CHOP)), antiapoptotic effects coupling with reduction of glial fibrillary acidic protein (GFAP) immunoreactivity and enhancement of phosphorylated c-AMP response element-binding (p-CREB) / brain-derived neurotrophic factor (BDNF)/tyrosine kinase B (TrkB) neuroprotective pathway. Briefly, Tirze exerts a promising role as adjuvant therapy in the colistin treatment protocol for protection against colistin's nephro- and neurotoxicity according to its anti-inflammatory, antioxidant, and antiapoptotic impacts besides its ability to suppress ER stress-related biomarkers.


Sujet(s)
Facteur neurotrophique dérivé du cerveau , Colistine , Protéine de liaison à l'élément de réponse à l'AMP cyclique , Stress du réticulum endoplasmique , Glycogen synthase kinase 3 beta , Rein , Stress oxydatif , Protéines proto-oncogènes c-akt , Transduction du signal , Animaux , Stress oxydatif/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Rats , Facteur neurotrophique dérivé du cerveau/métabolisme , Glycogen synthase kinase 3 beta/métabolisme , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Rein/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Rein/métabolisme , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Récepteur trkB/métabolisme , Neuroprotecteurs/usage thérapeutique , Neuroprotecteurs/pharmacologie , Rat Wistar , Antibactériens/usage thérapeutique , Antibactériens/effets indésirables , Syndromes neurotoxiques/traitement médicamenteux , Syndromes neurotoxiques/étiologie , Syndromes neurotoxiques/prévention et contrôle , Syndromes neurotoxiques/métabolisme , Maladies du rein/induit chimiquement , Maladies du rein/traitement médicamenteux , Maladies du rein/prévention et contrôle , Maladies du rein/métabolisme
19.
Ecotoxicol Environ Saf ; 279: 116497, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38805827

RÉSUMÉ

Methamphetamine (METH) is a highly abused substance on a global scale and has the capacity to elicit toxicity within the central nervous system. The neurotoxicity induced by METH encompasses neuronal degeneration and cellular demise within the substantia nigra-striatum and hippocampus. Caffeic acid phenethyl ester (CAPE), a constituent of propolis, is a diminutive compound that demonstrates antioxidative and anti-inflammatory characteristics. Numerous investigations have demonstrated the safeguarding effects of CAPE in various neurodegenerative ailments. Our hypothesis posits that CAPE may exert a neuroprotective influence on METH-induced neurotoxicity via specific mechanisms. In order to validate the hypothesis, a series of experimental techniques including behavioral tests, immunofluorescence labeling, RNA sequencing, and western blotting were employed to investigate the neurotoxic effects of METH and the potential protective effects of CAPE. The results of our study demonstrate that CAPE effectively ameliorates cognitive memory deficits and anxiety symptoms induced by METH in mice. Furthermore, CAPE has been observed to attenuate the upregulation of neurotoxicity-associated proteins that are induced by METH exposure and also reduced the loss of hippocampal neurons in mice. Moreover, transcriptomics analysis was conducted to determine alterations in gene expression within the hippocampus of mice. Subsequently, bioinformatics analysis was employed to investigate the divergent outcomes and identify potential key genes. Interferon-stimulated gene 15 (ISG15) was successfully identified and confirmed through RT-qPCR, western blotting, and immunofluorescence techniques. Our research findings unequivocally demonstrated the neuroprotective effect of CAPE against METH-induced neurotoxicity, with ISG15 may have an important role in the underlying protective mechanism. These results offer novel perspectives on the treatment of METH-induced neurotoxicity.


Sujet(s)
Acides caféiques , Métamfétamine , Neuroprotecteurs , Syndromes neurotoxiques , Alcool phénéthylique , Animaux , Acides caféiques/pharmacologie , Alcool phénéthylique/analogues et dérivés , Alcool phénéthylique/pharmacologie , Métamfétamine/toxicité , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Souris , Mâle , Syndromes neurotoxiques/prévention et contrôle , Syndromes neurotoxiques/traitement médicamenteux , Hippocampe/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Neurones/effets des médicaments et des substances chimiques
20.
Mol Biol Rep ; 51(1): 660, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38750264

RÉSUMÉ

BACKGROUND: Cadmium (Cd) is a heavy metal with extremely harmful toxic effects on the brain. Quetiapine (QTP) has unique neuroprotective effects with anti-inflammatory and antioxidant actions. However, its neuroprotective effect against Cd-induced neurotoxicity has not been previously studied. METHODS: QTP was administered in 10 and 20 mg/kg doses, while Cd was given in a dose of 6.5 mg/kg. RESULTS: In our study, QTP dose-dependently attenuated neuronal injury by downregulating p-tau and ß-amyloid. QTP potently attenuates histological abrasions induced by Cd. QTP counteracted oxidative injury by decreasing neuronal MDA and increased GSH levels mediated by downregulating Keap1 and upregulating Nrf2 and HO-1. QTP mitigated inflammation by decreasing MPO and NO2 and neuronal cytokines TNF-α and IL-1ß and upregulating IL-10 levels mediated by NF-κB downregulation. Additionally, QTP counteracted Cd-induced pyroptosis by downregulating caspase-1, ASC, and NLRP3 protein levels. CONCLUSION: In conclusion, QTP mitigates neurotoxicity induced by Cd through suppression of inflammation, pyroptosis, and oxidative stress by controlling the NF-κB, Keap1/Nrf2, and pyroptosis signals.


Sujet(s)
Cadmium , Inflammation , Stress oxydatif , Pyroptose , Fumarate de quétiapine , Stress oxydatif/effets des médicaments et des substances chimiques , Pyroptose/effets des médicaments et des substances chimiques , Animaux , Cadmium/toxicité , Fumarate de quétiapine/pharmacologie , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Mâle , Souris , Neuroprotecteurs/pharmacologie , Facteur-2 apparenté à NF-E2/métabolisme , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Neurones/anatomopathologie , Syndromes neurotoxiques/traitement médicamenteux , Syndromes neurotoxiques/métabolisme , Antioxydants/pharmacologie , Anti-inflammatoires/pharmacologie , Facteur de transcription NF-kappa B/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...