Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 812
Filtrer
1.
ACS Nano ; 18(36): 24872-24897, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-39197041

RÉSUMÉ

Potential exposure to cobalt nanoparticles (CoNPs) occurs in various fields, including hard alloy industrial production, the increasing use of new energy lithium-ion batteries, and millions of patients with metal-on-metal joint prostheses. Evidence from human, animal, and in vitro experiments suggests a close relationship between CoNPs and neurotoxicity. However, a systematic assessment of central nervous system (CNS) impairment due to CoNPs exposure and the underlying molecular mechanisms is lacking. In this study, we found that CoNPs induced neurodegenerative damage both in vivo and in vitro, including cognitive impairment, ß-amyloid deposition and Tau hyperphosphorylation. CoNPs promoted the formation of autophagosomes and impeding autophagosomal-lysosomal fusion in vivo and in vitro, leading to toxic protein accumulation. Moreover, CoNPs exposure reduced the level of transcription factor EB (TFEB) and the abundance of lysosome, causing a blockage in autophagosomal-lysosomal fusion. Interestingly, overexpression of long noncoding RNA NR_030777 mitigated CoNPs-induced neurodegenerative damage in both in vivo and in vitro models. Fluorescence in situ hybridization assay revealed that NR_030777 directly binds and stabilizes TFEB mRNA, alleviating the blockage of autophagosomal-lysosomal fusion and ultimately restoring neurodegeneration induced by CoNPs in vivo and in vitro. In summary, our study demonstrates that autophagic dysfunction is the main toxic mechanism of neurodegeneration upon CoNPs exposure and NR_030777 plays a crucial role in CoNPs-induced autophagic dysfunction. Additionally, the proposed adverse outcome pathway contributes to a better understanding of CNS toxicity assessment of CoNPs.


Sujet(s)
Autophagosomes , Cobalt , Lysosomes , Nanoparticules métalliques , ARN long non codant , Lysosomes/métabolisme , Lysosomes/effets des médicaments et des substances chimiques , ARN long non codant/génétique , ARN long non codant/métabolisme , Autophagosomes/métabolisme , Autophagosomes/effets des médicaments et des substances chimiques , Cobalt/composition chimique , Cobalt/pharmacologie , Animaux , Nanoparticules métalliques/composition chimique , Humains , Souris , Mâle , Autophagie/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Maladies neurodégénératives/métabolisme , Maladies neurodégénératives/anatomopathologie , Maladies neurodégénératives/induit chimiquement
2.
Neuroscience ; 557: 67-80, 2024 Oct 04.
Article de Anglais | MEDLINE | ID: mdl-39127344

RÉSUMÉ

Puberty is a sensitive developmental period during which stressors can cause lasting brain and behavioural deficits. While the acute effects of pubertal lipopolysaccharide (LPS) and antimicrobial (AMNS) treatments are known, their enduring impacts on neurodegeneration-related mechanisms and behaviours remain unclear. This study examined these effects in male and female mice. At five weeks old, mice received 200ul of either broad-spectrum antimicrobials or water through oral gavage twice daily for seven days. At six weeks of age, they received an intraperitoneal injection of either saline or LPS. Four weeks later, adult mice underwent neurodegeneration-related behavioural tests, including the rotarod, forepaw stride length, reversed grid hang, open field, and buried pellet tests. Two days after the final test, brain and ileal samples were collected. Results showed that female mice treated with both AMNS and LPS exhibited deficits in neuromuscular strength, while males treated with LPS alone showed increased anxiety-like behaviours. Males treated with AMNS alone had decreased sigma-1 receptor (S1R) expression in the cornu ammonis 1 (CA1) and dentate gyrus (DG), while females treated with both AMNS and LPS had decreased S1R expression. Additionally, males treated with either LPS or AMNS had lower glial-derived neurotrophic factor receptor alpha-1 (GFRA1) expression in the primary motor cortex (M1) than females. Mice treated with LPS alone had decreased GFRA1 expression in the DG and decreased S1R expression in the secondary motor cortex (M2). These findings suggest that pubertal AMNS and LPS treatments may lead to enduring changes in biomarkers and behaviours related to neurodegeneration.


Sujet(s)
Lipopolysaccharides , Animaux , Femelle , Mâle , Lipopolysaccharides/pharmacologie , Souris , Maturation sexuelle/effets des médicaments et des substances chimiques , Comportement animal/effets des médicaments et des substances chimiques , Maladies neurodégénératives/métabolisme , Maladies neurodégénératives/induit chimiquement , Maladies neurodégénératives/anatomopathologie , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Anxiété/induit chimiquement , Anxiété/métabolisme
3.
Ecotoxicol Environ Saf ; 284: 116960, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39208585

RÉSUMÉ

Accumulating observational studies have linked particulate air pollutants to neurodegenerative diseases (NDDs). However, the causal links and the direction of their associations remain unclear. Therefore, we adopted a two-sample Mendelian randomization (TSMR) design using the GWAS-based genetic instruments of particulate air pollutants (PM2.5 and PM10) from the UK Biobank to explore their causal influence on four common neurodegenerative diseases. Estimates of causative relationships were generated by the Inverse variance weighted (IVW) method with multiple sensitive analyses. The heterogeneity and pleiotropy tests were additionally performed to verify whether our findings were robust. Genetically predicted PM2.5 and PM10 could elevate the occurrence of AD (odds ratio [OR] = 2.22, 95 % confidence interval [CI] 1.53-3.22, PIVW = 2.85×10-5, PFalsediscovery rate[FDR]= 2.85×10-4 and OR = 2.41, 95 % CI: 1.26-4.60, PIVW = 0.008, PFDR=0.039, respectively). The results were robust in sensitive analysis. However, no evidence of causality was found for other NDDs. Our present study suggests that PM2.5 and PM10 have a detrimental effect on AD, which indicates that improving air quality to prevent AD may have pivotal public health implications.


Sujet(s)
Polluants atmosphériques , Analyse de randomisation mendélienne , Maladies neurodégénératives , Matière particulaire , Matière particulaire/analyse , Humains , Maladies neurodégénératives/génétique , Maladies neurodégénératives/induit chimiquement , Maladies neurodégénératives/épidémiologie , Polluants atmosphériques/analyse , Étude d'association pangénomique , Exposition environnementale/effets indésirables , Pollution de l'air/effets indésirables , Royaume-Uni
4.
Ecotoxicol Environ Saf ; 284: 116920, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39208581

RÉSUMÉ

Exposure to Particulate matter 2.5 (PM2.5) accelerates aging, causing declines in tissue and organ function, and leading to diseases such as cardiovascular, neurodegenerative, and musculoskeletal disorders. PM2.5 is a major environmental pollutant and an exogenous pathogen in air pollution that is now recognized as an accelerator of human aging and a predisposing factor for several age-related diseases. In this paper, we seek to elucidate the mechanisms by which PM2.5 induces cellular senescence, such as genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, and mitochondrial dysfunction, and age-related diseases. Our goal is to increase awareness among researchers within the field of the toxicity of environmental pollutants and to advocate for personal and public health initiatives to curb their production and enhance population protection. Through these endeavors, we aim to promote longevity and health in older adults.


Sujet(s)
Vieillissement , Polluants atmosphériques , Vieillissement de la cellule , Matière particulaire , Matière particulaire/toxicité , Humains , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Vieillissement/effets des médicaments et des substances chimiques , Polluants atmosphériques/toxicité , Instabilité du génome/effets des médicaments et des substances chimiques , Épigenèse génétique/effets des médicaments et des substances chimiques , Pollution de l'air/effets indésirables , Animaux , Exposition environnementale/effets indésirables , Mitochondries/effets des médicaments et des substances chimiques , Maladies neurodégénératives/induit chimiquement
5.
Chemosphere ; 364: 143180, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39187026

RÉSUMÉ

Cadmium (Cd) induced neurotoxicity has become a growing concern due to its potential adverse effects on the Central Nervous System. Cd is a Heavy Metal (HM) that is released into the environment, through several industrial processes. It poses a risk to the health of the community by polluting air, water, and soil. Cd builds up in the brain and other neural tissues, raising concerns about its effect on the nervous system due to its prolonged biological half-life. Cd can enter into the neurons, hence increasing the production of Reactive Oxygen Species (ROS) in them and impairing their antioxidant defenses. Cd disrupts the Calcium (Ca2+) balance in neurons, affects the function of the mitochondria, and triggers cell death pathways. As a result of these pathways, the path to the development of many neurological diseases affected by environmental factors, especially Cd, such as Alzheimer's Disease (AD) and Amyotrophic Lateral Sclerosis (ALS) is facilitated. There are cognitive deficits associated with long exposure to Cd. Memory disorders are present in both animals and humans. Cd alters the brain's function and performance in critical periods. There are lifelong consequences of Cd exposure during critical brain development stages. The susceptibility to neurotoxic effects is increased by interactions with a variety of risk factors. Cd poses risks to neuronal function and behavior, potentially contributing to neurodegenerative diseases like Parkinson's disease (PD) and AD as well as cognitive issues. This article offers a comprehensive overview of Cd-induced neurotoxicity, encompassing risk assessment, adverse effect levels, and illuminating intricate pathways.


Sujet(s)
Cadmium , Maladies neurodégénératives , Cadmium/toxicité , Humains , Maladies neurodégénératives/induit chimiquement , Animaux , Polluants environnementaux/toxicité , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Syndromes neurotoxiques/étiologie , Neurones/effets des médicaments et des substances chimiques
6.
Toxicol Lett ; 400: 35-41, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39117292

RÉSUMÉ

Lead (Pb2+) is one of the most common toxic metals present in the environment, and lead exposure causes serious health issues in humans. Lead is widely used because of its physio-chemical characteristics, which include softness, corrosion resistance, ductility, and low conductivity. Lead affects almost all human organs, specifically the central nervous system. Lead neurotoxicity is connected to various neural pathways, including brain-derived neurotrophic factor (BDNF) protein level alterations, cyclic adenosine 3',5'-monophosphate (cAMP) response element binding protein (CREB) pathway changes, and N-methyl-D-aspartate receptors (NMDARs) changes. Lead primarily affects protein kinase C (PKC) through the replacement of calcium (Ca2+) ions in the CREB pathway. In this review, we have discussed the effect of lead on the CREB pathway and its implications on the nervous system, highlighting its effects on learning, synaptic plasticity, memory, and cognitive deficits. This review provides an understanding of the lead-induced alterations in the CREB pathway, which can lead to the future prospect of its use as a diagnostic marker as well as a therapeutic target for neurodegenerative disorders.


Sujet(s)
Protéine de liaison à l'élément de réponse à l'AMP cyclique , Plomb , Maladies neurodégénératives , Humains , Plomb/toxicité , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Maladies neurodégénératives/métabolisme , Maladies neurodégénératives/induit chimiquement , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Plasticité neuronale/effets des médicaments et des substances chimiques
7.
STAR Protoc ; 5(3): 103243, 2024 Sep 20.
Article de Anglais | MEDLINE | ID: mdl-39096497

RÉSUMÉ

In vivo models of brain pathology are crucial for studying neurological diseases. Here, we present a protocol to induce a pathological condition in a mouse brain area by local injection of neurotoxic stimulus. We describe steps for preparing reagents, stereotaxic injection procedures to induce neurodegeneration in the hippocampus, and preparation of brain sections to examine the induced model. This protocol is useful for studying how local pathology affects other brain areas and neighbor cells and its functional consequences in behavior. For complete details on the use and execution of this protocol, please refer to Zhang et al.1.


Sujet(s)
Encéphale , Techniques stéréotaxiques , Animaux , Souris , Encéphale/effets des médicaments et des substances chimiques , Encéphale/anatomopathologie , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/anatomopathologie , Maladies neurodégénératives/induit chimiquement , Maladies neurodégénératives/anatomopathologie , Modèles animaux de maladie humaine
8.
Environ Int ; 190: 108897, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39047545

RÉSUMÉ

The utilization of Cobalt (Co) has surged due to it is critical role in renewable energy technologies and other high-tech applications. Concurrently, the potential health risks associated with Co exposure have raised concerns. Previous studies, including our own, have shown that Co can impair learn and memory functions as an epigenetic hazard, even at low concentrations. In this study, we explore the mechanisms of Co-induced ferroptosis in neurodegenerative damage both in vivo and in vitro, focusing on the epigenetic regulation by N6-methyladenosine (m6A) demethylase alkB homolog 5 (ALKBH5). We identify heme oxygenase-1 (HO-1) as a direct target gene of ALKBH5, playing a crucial role in mitigating Co-induced ferroptosis. ALKBH5 deficiency affects the post-transcriptional regulation of HO-1 through m6A modification, which in turn influences mRNA's stability, intracellular distribution, and alternative splicing, thereby enhancing susceptibility to Co-induced ferroptosis. Additionally, we discuss the potential involvement of heterogeneous nuclear ribonucleoprotein M (hnRNPM) in regulating alternative splicing of HO-1 mRNA, potentially mediated by m6A modifications. This study provides new epigenetic insights into the post-transcriptional regulatory mechanisms involved in Co-induced ferroptosis and highlights the broader implications of environmental hazards in neurodegenerative damage.


Sujet(s)
Adénosine , AlkB Homolog 5, RNA demethylase , Cobalt , Ferroptose , Heme oxygenase-1 , ARN messager , Heme oxygenase-1/génétique , Heme oxygenase-1/métabolisme , AlkB Homolog 5, RNA demethylase/métabolisme , AlkB Homolog 5, RNA demethylase/génétique , Animaux , Ferroptose/effets des médicaments et des substances chimiques , Adénosine/analogues et dérivés , Adénosine/métabolisme , Cobalt/toxicité , ARN messager/génétique , ARN messager/métabolisme , Souris , Humains , Maladies neurodégénératives/induit chimiquement , Maladies neurodégénératives/génétique , Épigenèse génétique
9.
Environ Res ; 259: 119552, 2024 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-38964584

RÉSUMÉ

BACKGROUND: Long-term exposure to ambient air pollution has been linked with all-cause mortality and cardiovascular and respiratory diseases. Suggestive associations between ambient air pollutants and neurodegeneration have also been reported, but due to the small effect and relatively rare outcomes evidence is yet inconclusive. Our aim was to investigate the associations between long-term air pollution exposure and mortality from neurodegenerative diseases. METHODS: A Dutch national cohort of 10.8 million adults aged ≥30 years was followed from 2013 until 2019. Annual average concentrations of air pollutants (ultra-fine particles (UFP), nitrogen dioxide (NO2), fine particles (PM2.5 and PM10) and elemental carbon (EC)) were estimated at the home address at baseline, using land-use regression models. The outcome variables were mortality due to amyotrophic lateral sclerosis (ALS), Parkinson's disease, non-vascular dementia, Alzheimer's disease, and multiple sclerosis (MS). Hazard ratios (HR) were estimated using Cox models, adjusting for individual and area-level socio-economic status covariates. RESULTS: We had a follow-up of 71 million person-years. The adjusted HRs for non-vascular dementia were significantly increased for NO2 (1.03; 95% confidence interval (CI) 1.02-1.05) and PM2.5 (1.02; 95%CI 1.01-1.03) per interquartile range (IQR; 6.52 and 1.47 µg/m3, respectively). The association with PM2.5 was also positive for ALS (1.02; 95%CI 0.97-1.07). These associations remained positive in sensitivity analyses and two-pollutant models. UFP was not associated with any outcome. No association with air pollution was found for Parkinson's disease and MS. Inverse associations were found for Alzheimer's disease. CONCLUSION: Our findings, using a cohort of more than 10 million people, provide further support for associations between long-term exposure to air pollutants (PM2.5 and particularly NO2) and mortality of non-vascular dementia. No associations were found for Parkinson and MS and an inverse association was observed for Alzheimer's disease.


Sujet(s)
Polluants atmosphériques , Pollution de l'air , Exposition environnementale , Maladies neurodégénératives , Matière particulaire , Humains , Pays-Bas/épidémiologie , Pollution de l'air/effets indésirables , Pollution de l'air/analyse , Mâle , Polluants atmosphériques/analyse , Polluants atmosphériques/effets indésirables , Maladies neurodégénératives/mortalité , Maladies neurodégénératives/induit chimiquement , Maladies neurodégénératives/épidémiologie , Adulte d'âge moyen , Femelle , Études de cohortes , Exposition environnementale/effets indésirables , Sujet âgé , Matière particulaire/analyse , Matière particulaire/effets indésirables , Adulte
10.
Toxicol Sci ; 201(2): 254-262, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39067045

RÉSUMÉ

The mechanisms associated with neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD), have yet to be fully characterized, and genetic as well as environmental factors in their disease etiology are underappreciated. Although mutations in genes such as PARKIN and LRRK2 have been linked to PD, the idiopathic component of the disease suggests a contribution of environmental risk factors, including metals, such as copper (Cu). Cu overexposure has been reported to cause oxidative stress and neurotoxicity, but its role in neurodegenerative diseases is rarely studied. Using Caenorhabditis elegans (C. elegans) as a model organism for neurotoxicity, we assessed the effects of Cu oversupply in AD and PD models. Our findings reveal that although copper treatment did not induce neurodegeneration in wild-type worms or the AD model, it significantly exacerbated neurodegeneration in the PD-associated mutants PARKIN and LRRK2. These results suggest that genetic predisposition for PD enhances the sensitivity to copper toxicity, highlighting the multifactorial nature of neurodegenerative diseases. Furthermore, our study provides insight into the mechanisms underlying Cu-induced neurotoxicity in PD models, including disruptions in dopamine levels, altered dopamine-dependent behavior and degraded dopaminergic neurons. Overall, our novel findings contribute to a better understanding of the complex interactions between genetic susceptibility, environmental factors, and neurodegenerative disease pathogenesis, emphasizing the importance of a tightly regulated Cu homeostasis in the etiology of PD. Copper oversupply exacerbated neurodegeneration in Caenorhabditis elegans models of Parkinson's disease, highlighting the genetic susceptibility and emphasizing the crucial role of tightly regulated copper homeostasis in Parkinson's disease pathogenesis.


Sujet(s)
Caenorhabditis elegans , Cuivre , Caenorhabditis elegans/génétique , Caenorhabditis elegans/effets des médicaments et des substances chimiques , Animaux , Cuivre/toxicité , Protéines de Caenorhabditis elegans/génétique , Protéines de Caenorhabditis elegans/métabolisme , Maladie de Parkinson/génétique , Maladie de Parkinson/étiologie , Maladies neurodégénératives/induit chimiquement , Maladies neurodégénératives/génétique , Ubiquitin-protein ligases/génétique , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/induit chimiquement , Leucine-rich repeat serine-threonine protein kinase-2/génétique , Mutation , Animal génétiquement modifié , Prédisposition génétique à une maladie , Modèles animaux de maladie humaine
11.
Neuroreport ; 35(13): 832-838, 2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-38973498

RÉSUMÉ

This study aimed to investigate the effects of rolipram, a phosphodiesterase inhibitor, on brain tissue regeneration. Trimethyltin-injected mice, an animal model of hippocampal tissue regeneration, was created by a single injection of trimethyltin chloride (2.2 mg/kg, intraperitoneally). Daily rolipram administration (10 mg/kg, intraperitoneally) was performed from the day after trimethyltin injection until the day before sampling. In Experiment 1, brain samples were collected on day 7 postinjection of trimethyltin following the forced swim test. In Experiment 2, bromodeoxyuridine (150 mg/kg, intraperitoneally/day) was administered on days 3-5 and sampling was on day 21 postinjection of trimethyltin. Samples were routinely embedded in paraffin and sections were obtained for histopathological investigation. In Experiment 1, rolipram-treated mice showed shortened immobility times in the forced swim test. Histopathology revealed that rolipram treatment had improved the replenishment of neuronal nuclei-positive neurons in the dentate gyrus, which was accompanied by an increase in the percentage of phosphorylated cyclic AMP response element-binding protein-positive cells. In addition, rolipram had decreased the percentage of ionized calcium-binding adapter protein 1-positive microglia with activated morphology and the number of tumor necrosis factor-alpha-expressing cells. In Experiment 2, double immunofluorescence for bromodeoxyuridine/neuronal nuclei revealed an increase of double-positive cells in rolipram-treated mice. These results demonstrate that rolipram effectively promotes brain tissue regeneration by enhancing the survival of newborn neurons and inhibiting neuroinflammation.


Sujet(s)
Hippocampe , Rolipram , Triméthyl-stannanes , Animaux , Triméthyl-stannanes/toxicité , Rolipram/pharmacologie , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/anatomopathologie , Hippocampe/métabolisme , Souris , Mâle , Maladies neurodégénératives/induit chimiquement , Maladies neurodégénératives/traitement médicamenteux , Maladies neurodégénératives/anatomopathologie , Régénération nerveuse/effets des médicaments et des substances chimiques , Régénération nerveuse/physiologie , Neurones/effets des médicaments et des substances chimiques , Neurones/anatomopathologie , Microglie/effets des médicaments et des substances chimiques , Microglie/anatomopathologie
12.
BMC Med ; 22(1): 266, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38951846

RÉSUMÉ

BACKGROUND: Benzodiazepine use is common, particularly in older adults. Benzodiazepines have well-established acute adverse effects on cognition, but long-term effects on neurodegeneration and dementia risk remain uncertain. METHODS: We included 5443 cognitively healthy (MMSE ≥ 26) participants from the population-based Rotterdam Study (57.4% women, mean age 70.6 years). Benzodiazepine use from 1991 until baseline (2005-2008) was derived from pharmacy dispensing records, from which we determined drug type and cumulative dose. Benzodiazepine use was defined as prescription of anxiolytics (ATC-code: N05BA) or sedative-hypnotics (ATC-code: N05CD) between inception of pharmacy records and study baseline. Cumulative dose was calculated as the sum of the defined daily doses for all prescriptions. We determined the association with dementia risk until 2020 using Cox regression. Among 4836 participants with repeated brain MRI, we further determined the association of benzodiazepine use with changes in neuroimaging markers using linear mixed models. RESULTS: Of all 5443 participants, 2697 (49.5%) had used benzodiazepines at any time in the 15 years preceding baseline, of whom 1263 (46.8%) used anxiolytics, 530 (19.7%) sedative-hypnotics, and 904 (33.5%) used both; 345 (12.8%) participants were still using at baseline assessment. During a mean follow-up of 11.2 years, 726 participants (13.3%) developed dementia. Overall, use of benzodiazepines was not associated with dementia risk compared to never use (HR [95% CI]: 1.06 [0.90-1.25]), irrespective of cumulative dose. Risk estimates were somewhat higher for any use of anxiolytics than for sedative-hypnotics (HR 1.17 [0.96-1.41] vs 0.92 [0.70-1.21]), with strongest associations for high cumulative dose of anxiolytics (HR [95% CI] 1.33 [1.04-1.71]). In imaging analyses, current use of benzodiazepine was associated cross-sectionally with lower brain volumes of the hippocampus, amygdala, and thalamus and longitudinally with accelerated volume loss of the hippocampus and to a lesser extent amygdala. However, imaging findings did not differ by type of benzodiazepines or cumulative dose. CONCLUSIONS: In this population-based sample of cognitively healthy adults, overall use of benzodiazepines was not associated with increased dementia risk, but potential class-dependent adverse effects and associations with subclinical markers of neurodegeneration may warrant further investigation.


Sujet(s)
Benzodiazépines , Démence , Humains , Femelle , Démence/épidémiologie , Démence/induit chimiquement , Mâle , Sujet âgé , Benzodiazépines/effets indésirables , Benzodiazépines/administration et posologie , Adulte d'âge moyen , Imagerie par résonance magnétique , Pays-Bas/épidémiologie , Sujet âgé de 80 ans ou plus , Neuroimagerie , Encéphale/imagerie diagnostique , Encéphale/effets des médicaments et des substances chimiques , Encéphale/anatomopathologie , Études prospectives , Maladies neurodégénératives/épidémiologie , Maladies neurodégénératives/induit chimiquement , Hypnotiques et sédatifs/effets indésirables , Facteurs de risque
13.
J Neuroimmunol ; 392: 578374, 2024 07 15.
Article de Anglais | MEDLINE | ID: mdl-38797060

RÉSUMÉ

We aimed to investigate ampicillin (AMP) mechanisms in microbiota-gut-brain axis. We evaluated its effect on two gut and brain regions and behavioral performances. We administred AMP (1 g/l) to BALB/c mice for 21 days. Then, we analyzed body weigth change, stool consistency scoring, gut length, intestinal microbiota composition, nitric oxide synthase 2 (NOS2) expression and tissue integrity. We subsequently evaluated NOS2, GFAP, CD68 and NFL cerebral expression and spatial memory.Interestingly, our data showed gut microbiota disruption, NOS2 upregulation and tissue damage, associated to cerebral NOS2, GFAP, CD68 and NFL over-expression and behavioral alteration. Antiobiotic therapy should be prescribed with great caution.


Sujet(s)
Ampicilline , Axe cerveau-intestin , Dysbiose , Microbiome gastro-intestinal , Souris de lignée BALB C , Nitric oxide synthase type II , Animaux , Souris , Ampicilline/pharmacologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Microbiome gastro-intestinal/physiologie , Dysbiose/induit chimiquement , Nitric oxide synthase type II/métabolisme , Mâle , Axe cerveau-intestin/physiologie , Axe cerveau-intestin/effets des médicaments et des substances chimiques , Maladies neuro-inflammatoires/métabolisme , Antibactériens/pharmacologie , Mémoire spatiale/effets des médicaments et des substances chimiques , Mémoire spatiale/physiologie , Modèles animaux de maladie humaine , Maladies neurodégénératives/induit chimiquement
14.
Toxicol Mech Methods ; 34(7): 781-794, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38699799

RÉSUMÉ

Bisphenol A (BPA), a common plasticizer, is categorized as a neurotoxic compound. Its impact on individuals exhibits sex-linked variations. Several biological and environmental factors impact the degree of toxicity. Moreover, nutritional factors have profound influence on toxicity outcome. BPA has been demonstrated to be an obesogen. However, research on the potential role of obesity as a confounding factor in BPA toxicity is lacking. We studied the neurodegenerative effects in high-fat diet (HFD)-induced obese female rats after exposure to BPA (10 mg/L via drinking water for 90 days). Four groups were taken in this study - Control, HFD, HFD + BPA and BPA. Cognitive function was evaluated through novel object recognition (NOR) test. Inflammatory changes in brain, and changes in hormonal level, lipid profile, glucose tolerance, oxidative stress, and antioxidants were also determined. HFD + BPA group rats showed a significant decline in memory function in NOR test. The cerebral cortex (CC) of the brain showed increased neurodegenerative changes as measured by microtubule-associated protein-2 (MAP-2) accompanied by histopathological confirmation. The increased level of neuroinflammation was demonstrated by microglial activation (Iba-1) and protein expression of nuclear factor- kappa B (NF-КB) in the brain. Obesity also caused significant (p < 0.05) increase in lipid peroxidation accompanied by reduced activities of antioxidant enzymes (glutathione S-transferase, catalase and glutathione peroxidase) and decrease in reduced-glutathione (p < 0.05) when compared to non-obese rats with BPA treatment. Overall, study revealed that obesity serves as a risk factor in the toxicity of BPA which may exacerbate the progression of neurological diseases.


Sujet(s)
Composés benzhydryliques , Alimentation riche en graisse , Maladies neuro-inflammatoires , Obésité , Phénols , Animaux , Composés benzhydryliques/toxicité , Phénols/toxicité , Femelle , Obésité/induit chimiquement , Obésité/métabolisme , Maladies neuro-inflammatoires/induit chimiquement , Maladies neuro-inflammatoires/anatomopathologie , Stress oxydatif/effets des médicaments et des substances chimiques , Rat Wistar , Rats , Peroxydation lipidique/effets des médicaments et des substances chimiques , Maladies neurodégénératives/induit chimiquement , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Encéphale/anatomopathologie
15.
Environ Toxicol ; 39(9): 4267-4277, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38700421

RÉSUMÉ

Neurodegeneration in conditions like Alzheimer's and Parkinson's disease is influenced by genetic and environmental factors. This study explores the potential neurodegenerative effects of lead (Pb) toxicity and amyloid beta peptides (Aßp 1-40 and Aßp 25-35) by promoting M1 polarization in microglial cells. To this end, we investigated and observed that IC50 concentrations of Pb (22.8 µM) and Aßp 25-35(29.6 µM). Our results demonstrated significant Pb uptake (31.13% at 25 µM Pb) and increased intracellular ROS levels (77.1%) upon treatment with Pb in combination of both Aßp 1-40 and Aßp 25-35. Protein carbonylation significantly increased (73.12 nmol/mL) upon treatment with Pb in combination of both Aßp 1-40 and Aßp 25-35, indicating oxidative damage and compromised cellular defenses against oxidative stress along with elevated DNA oxidative damage (164.9 pg/mL of 8-OH-dG) upon treatment with Pb in combination with both Aßp 1-40 and Aßp 25-35. Microglial polarization showed elevated M1 markers (inducible nitric oxide synthase and cyclooxygenase 2) and reduced M2 markers (arginase-1 and cluster of differentiation 206), suggesting Pb's role in inducing neurodegenerative microglial polarization. These findings provide insights into the complex molecular events contributing to Pb-induced neurotoxicity and neurodegenerative diseases.


Sujet(s)
Peptides bêta-amyloïdes , Plomb , Microglie , Stress oxydatif , Espèces réactives de l'oxygène , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Peptides bêta-amyloïdes/toxicité , Peptides bêta-amyloïdes/métabolisme , Animaux , Plomb/toxicité , Stress oxydatif/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Nitric oxide synthase type II/métabolisme , Fragments peptidiques/toxicité , Fragments peptidiques/métabolisme , Cyclooxygenase 2/métabolisme , Souris , Lignée cellulaire , Altération de l'ADN/effets des médicaments et des substances chimiques , Maladies neurodégénératives/induit chimiquement
16.
Tissue Cell ; 88: 102393, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38705086

RÉSUMÉ

BACKGROUND: The cognitive deficits observed after treatment with chemotherapeutic drugs are obvious clinical problems. For treating chemotherapy-induced cognitive deficits (CICD), the treatment modalities must target its underlying mechanisms. Specifically, cisplatin may activate glycogen synthase kinase-3ß (GSK-3ß), thereby enhancing neuronal apoptosis. 6-bromoindirubin-3'-oxime (6BIO) was not investigated previously in a model of CICD. Therefore, this investigation aimed to address the impacts of GSK3 inhibition on regulating cell signaling, which contributes to neurodegeneration and cognitive impairment. METHODS: Thirty adult male Wistar rats were randomly allocated into control groups, while two experimental groups were exposed to repeated cisplatin injections (2 mg/kg intraperitoneally (ip), twice weekly, nine injections), termed chemobrain groups. The rats in the two experimental groups were equally divided into the chemobrain group (untreated) and the chemobrain-6BIO group (treated with 6BIO at a dose of 8.5 µg/kg ip every two days, started after the last dose of cisplatin and continued for two weeks). RESULTS: Repeated exposure to cisplatin led to a marked decline in cognitive functions. GSK3 inhibition exerted neuroprotection by decreasing the expression of p-tau and amyloid ß, thereby improving cognition. 6BIO, the GSK-3ß inhibitor, restored mitochondrial biogenesis by augmenting the protein levels of PGC1-α and increasing the number of mitochondria in the cerebral cortex and hippocampus. CONCLUSION: 6BIO provided neuroprotection and exhibited anti-apoptotic and anti-oxidative effects in a rat model of chemobrain.


Sujet(s)
Cisplatine , Glycogen synthase kinase 3 beta , Indoles , Biogenèse des organelles , Oximes , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes , Rat Wistar , Animaux , Oximes/pharmacologie , Glycogen synthase kinase 3 beta/métabolisme , Indoles/pharmacologie , Cisplatine/pharmacologie , Mâle , Rats , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Maladies neurodégénératives/traitement médicamenteux , Maladies neurodégénératives/métabolisme , Maladies neurodégénératives/induit chimiquement
17.
J Biochem Mol Toxicol ; 38(5): e23717, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38742857

RÉSUMÉ

Aluminum chloride (AlCl3) is a potent neurotoxic substance known to cause memory impairment and oxidative stress-dependent neurodegeneration. Naringenin (NAR) is a dietary flavonoid with potent antioxidant and anti-inflammatory properties which was implemented against AlCl3-induced neurotoxicity to ascertain its neuroprotective efficacy. Experimental neurotoxicity in mice was induced by exposure of AlCl3 (10 mg/kg, p.o.) followed by treatment with NAR (10 mg/kg, p.o.) for a total of 63 days. Assessed the morphometric, learning memory dysfunction (novel object recognition, T- and Y-maze tests), neuronal oxidative stress, and histopathological alteration in different regions of the brain, mainly cortex, hippocampus, thalamus, and cerebellum. AlCl3 significantly suppressed the spatial learning and memory power which were notably improved by administration of NAR. The levels of oxidative stress parameters nitric oxide, advanced oxidation of protein products, protein carbonylation, lipid peroxidation, superoxide dismutase, catalase, glutathione reductase, reduced glutathione, and the activity of acetylcholine esterase were altered 1.5-3 folds by AlCl3 significantly. Treatment of NAR remarkably restored the level of oxidative stress parameters and maintained the antioxidant defense system. AlCl3 suppressed the expression of neuronal proliferation marker NeuN that was restored by NAR treatment which may be a plausible mechanism. NAR showed therapeutic efficacy as a natural supplement against aluminum-intoxicated memory impairments and histopathological alteration through a mechanism involving an antioxidant defense system and neuronal proliferation.


Sujet(s)
Chlorure d'aluminium , Flavanones , Troubles de la mémoire , Stress oxydatif , Animaux , Flavanones/pharmacologie , Flavanones/usage thérapeutique , Stress oxydatif/effets des médicaments et des substances chimiques , Souris , Troubles de la mémoire/induit chimiquement , Troubles de la mémoire/traitement médicamenteux , Troubles de la mémoire/métabolisme , Chlorure d'aluminium/toxicité , Mâle , Maladies neurodégénératives/induit chimiquement , Maladies neurodégénératives/traitement médicamenteux , Maladies neurodégénératives/métabolisme , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Encéphale/anatomopathologie , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique
18.
Sci Total Environ ; 933: 173041, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38723972

RÉSUMÉ

Although many organochlorine pesticides (OCPs) have been banned or restricted because of their persistence and linkage to neurodegenerative diseases, there is evidence of continued human exposure. In contrast, registered herbicides are reported to have a moderate to low level of toxicity; however, there is little information regarding their toxicity to humans or their combined effects with OCPs. This study aimed to characterize the mechanism of toxicity of banned OCP insecticides (aldrin, dieldrin, heptachlor, and lindane) and registered herbicides (trifluralin, triallate, and clopyralid) detected at a legacy contaminated pesticide manufacturing and packing site using SH-SY5Y cells. Cell viability, LDH release, production of reactive oxygen species (ROS), and caspase 3/7 activity were evaluated following 24 h of exposure to the biocides. In addition, RNASeq was conducted at sublethal concentrations to investigate potential mechanisms involved in cellular toxicity. Our findings suggested that aldrin and heptachlor were the most toxic, while dieldrin, lindane, trifluralin, and triallate exhibited moderate toxicity, and clopyralid was not toxic to SH-SY5Y cells. While aldrin and heptachlor induced their toxicity through damage to the cell membrane, the toxicity of dieldrin was partially attributed to necrosis and apoptosis. Moreover, toxic effects of lindane, trifluralin, and triallate, at least partially, were associated with ROS generation. Gene expression profiles suggested that decreased cell viability induced by most of the tested biocides was related to inhibited cell proliferation. The dysregulation of genes encoding for proteins with anti-apoptotic properties also supported the absence of caspase activation. Identified enriched terms showed that OCP toxicity in SH-SY5Y cells was mediated through pathways associated with the pathogenesis of neurodegenerative diseases. In conclusion, this study provides a basis for elucidating the molecular mechanisms of pesticide-induced neurotoxicity. Moreover, it introduced SH-SY5Y cells as a relevant in vitro model for investigating the neurotoxicity of pesticides in humans.


Sujet(s)
Neuroblastome , Maladies neurodégénératives , Espèces réactives de l'oxygène , Humains , Maladies neurodégénératives/induit chimiquement , Lignée cellulaire tumorale , Espèces réactives de l'oxygène/métabolisme , Pesticides/toxicité , Dieldrine/toxicité , Insecticides/toxicité , Survie cellulaire/effets des médicaments et des substances chimiques , Hydrocarbures chlorés/toxicité , Apoptose/effets des médicaments et des substances chimiques , Herbicides/toxicité , Aldrine/toxicité , Lindane/toxicité
19.
MAGMA ; 37(5): 765-790, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-38635150

RÉSUMÉ

Neurodegenerative disorders, including Multiple Sclerosis (MS), are heterogenous disorders which affect the myelin sheath of the central nervous system (CNS). Magnetic Resonance Imaging (MRI) provides a non-invasive method for studying, diagnosing, and monitoring disease progression. As an emerging research area, many studies have attempted to connect MR metrics to underlying pathophysiological presentations of heterogenous neurodegeneration. Most commonly, small animal models are used, including Experimental Autoimmune Encephalomyelitis (EAE), Theiler's Murine Encephalomyelitis (TMEV), and toxin models including cuprizone (CPZ), lysolecithin, and ethidium bromide (EtBr). A contrast and comparison of these models is presented, with focus on the cuprizone model, followed by a review of literature studying neurodegeneration using MRI and the cuprizone model. Conventional MRI methods including T1 Weighted (T1W) and T2 Weighted (T2W) Imaging are mentioned. Quantitative MRI methods which are sensitive to diffusion, magnetization transfer, susceptibility, relaxation, and chemical composition are discussed in relation to studying the CPZ model. Overall, additional studies are needed to improve both the sensitivity and specificity of MRI metrics for underlying pathophysiology of neurodegeneration and the relationships in attempts to clear the clinico-radiological paradox. We therefore propose a multiparametric approach for the investigation of MR metrics for underlying pathophysiology.


Sujet(s)
Cuprizone , Maladies démyélinisantes , Modèles animaux de maladie humaine , Imagerie par résonance magnétique , Cuprizone/toxicité , Animaux , Souris , Imagerie par résonance magnétique/méthodes , Maladies démyélinisantes/imagerie diagnostique , Maladies démyélinisantes/induit chimiquement , Maladies neurodégénératives/imagerie diagnostique , Maladies neurodégénératives/induit chimiquement , Sclérose en plaques/imagerie diagnostique , Sclérose en plaques/induit chimiquement , Gaine de myéline , Encéphalomyélite auto-immune expérimentale/imagerie diagnostique , Encéphalomyélite auto-immune expérimentale/induit chimiquement , Humains , Sensibilité et spécificité
20.
Environ Int ; 186: 108597, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38579453

RÉSUMÉ

The growing body of evidence links exposure to particulate matter pollutants with an increased risk of neurodegenerative diseases. In the present study, we investigated whether diesel exhaust particles can induce neurobehavioral alterations associated with neurodegenerative effects on glutamatergic and dopaminergic neurons in Caenorhabditis elegans (C. elegans). Exposure to DEP at concentrations of 0.167 µg/cm2 and 1.67 µg/cm2 resulted in significant developmental delays and altered locomotion behaviour. These effects were accompanied by discernible alterations in the expressions of antioxidant genes sod-3 and gst-4 observed in transgenic strains. Behaviour analysis demonstrated a significant reduction in average speed (p < 0.001), altered paths, and decreased swimming activities (p < 0.01), particularly at mid and high doses. Subsequent assessment of neurodegeneration markers in glutamatergic (DA1240) and dopaminergic (BZ555) transgenic worms revealed notable glutamatergic neuron degeneration at 0.167 µg/cm2 (∼30 % moderate, ∼20 % advanced) and 1.67 µg/cm2 (∼28 % moderate, ∼24 % advanced, p < 0.0001), while dopaminergic neurons exhibited structural deformities (∼16 %) without significant degeneration in terms of blebs and breaks. Furthermore, in silico docking simulations suggest the presence of an antagonistic competitive inhibition induced by DEP in the evaluated neuro-targets, stronger for the glutamatergic transporter than for the dopaminergic receptor from the comparative binding affinity point of view. The results underscore DEP's distinctive neurodegenerative effects and suggest a link between locomotion defects and glutamatergic neurodegeneration in C. elegans, providing insights into environmental health risks assessment.


Sujet(s)
Caenorhabditis elegans , Neurones dopaminergiques , Emissions des véhicules , Animaux , Caenorhabditis elegans/effets des médicaments et des substances chimiques , Neurones dopaminergiques/effets des médicaments et des substances chimiques , Emissions des véhicules/toxicité , Matière particulaire/toxicité , Animal génétiquement modifié , Acide glutamique/métabolisme , Locomotion/effets des médicaments et des substances chimiques , Maladies neurodégénératives/induit chimiquement , Polluants atmosphériques/toxicité
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE