Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.411
Filtrer
1.
CNS Neurosci Ther ; 30(10): e70068, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39392360

RÉSUMÉ

BACKGROUND: Alzheimer's disease (AD) is a common chronic neurodegenerative disease in older people, and there is no specific treatment that can stop or reverse its progression. Neobavaisoflavone (NBIF) is a flavonoid that has been shown to have neuroprotective effects, but its role in AD has not been revealed. The present study investigated the role and mechanism of NBIF on Aß25-35-induced brain injury. METHODS: In this experiment, the AD mouse model was established by injection of Aß25-35 peptides (200 µM, icv), and Donepezil (Don, 10 mg/kg/days), NBIF-L (15 mg/kg/days), and NBIF-H (30 mg/kg/days) were administered orally for 4 weeks. Learning memory, hippocampal pathological changes, pathological markers, apoptosis, oxidative stress, inflammation, immune cells were measured in mice. Network pharmacology combined with the GEO database led to the identification of SIRT1, a key target for NBIF intervention in AD, and levels of SIRT1, p-STAT3 and FOXO1 were measured. In addition, the antagonistic activity of SIRT1 transfection silencing against NBIF in Aß25-35-induced in N9 cells and N2a-APP69 cells was investigated to assess whether the effects caused by NBIF were mediated by SIRT1. RESULTS: The results showed that NBIF ameliorated learning memory and hippocampal neuronal damage, reduced pathological markers, apoptosis, oxidative stress and neuroinflammation, and modulated immune cells. SIRT1 is a key target for NBIF intervention in AD, and NBIF upregulates SIRT1 and reduces the expression levels of p-STAT3 and FOXO1. Furthermore, silencing SIRT1 effectively reduced the protective effect of NBIF on Aß25-35-induced N9 cells and N2a-APP69 cells, which indicated that the protective effect of NBIF on AD is related to SIRT1. CONCLUSIONS: NBIF ameliorated Aß25-35-induced brain injury by inhibiting apoptosis, oxidative stress, and neuroinflammation, which may be mediated through SIRT1 signaling. These findings provide a rationale for NBIF in the treatment of AD and help facilitate the development of clinical therapeutic agents for AD.


Sujet(s)
Peptides bêta-amyloïdes , Troubles de la mémoire , Fragments peptidiques , Sirtuine-1 , Animaux , Sirtuine-1/métabolisme , Troubles de la mémoire/induit chimiquement , Troubles de la mémoire/traitement médicamenteux , Peptides bêta-amyloïdes/toxicité , Souris , Fragments peptidiques/toxicité , Mâle , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Souris de lignée C57BL , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/anatomopathologie , Hippocampe/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Flavonoïdes/pharmacologie , Flavonoïdes/usage thérapeutique , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques , Maladie d'Alzheimer/induit chimiquement , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/métabolisme , Apoptose/effets des médicaments et des substances chimiques
2.
Noise Health ; 26(122): 396-402, 2024.
Article de Anglais | MEDLINE | ID: mdl-39345083

RÉSUMÉ

The developing brain is significantly affected by long-term exposure to noise at an early age, leading to functional disorders such as learning and memory impairments. Gastrodin (GAS), a natural organic compound, is an extraction of phenolic glycoside from the rhizome of Gastrodia elata. Clinically, GAS is extensively utilised for the treatment of neurological disorders. This study aimed to explore the effect and mechanism of GAS on noise exposure-induced learning and memory impairments. Rats aged 21 days were exposed to a 90 dB noise environment for 4 weeks and divided into the noise group, the noise + GAS group, and the control group to establish a noise exposure model. After noise exposure treatment, the improvement effect of GAS on the memory of rats was evaluated by Y-maze and Morris water maze. Enzyme-linked immunosorbent assay was utilised to determine the effect of GAS on neurotransmitter levels in the hippocampal tissue of noise-exposed rats. Western blot was applied for the detection of the protein levels of neurotrophic factors. The GAS treatment significantly improved spatial memory and increased the levels of key neurotransmitters (norepinephrine, dopamine and serotonin) and neurotrophic factors (neurotrophin-3 and brain-derived neurotrophic factor) in the hippocampal tissues of noise-exposed rats. These alterations correlate with enhanced cognitive functions, suggesting a neuroprotective effect of GAS against noise-induced cognitive impairments. This study supports the potential of GAS to treat noise-induced learning and memory impairments by modulating neurotransmitter secretion and enhancing the expression levels of neurotrophic factors. These findings offer potential therapeutic avenues for cognitive impairments induced by noise exposure.


Sujet(s)
Alcools benzyliques , Glucosides , Hippocampe , Apprentissage du labyrinthe , Troubles de la mémoire , Bruit , Animaux , Glucosides/pharmacologie , Rats , Alcools benzyliques/pharmacologie , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Troubles de la mémoire/étiologie , Troubles de la mémoire/traitement médicamenteux , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques , Mâle , Bruit/effets indésirables , Rat Sprague-Dawley , Facteur neurotrophique dérivé du cerveau/métabolisme , Neuroprotecteurs/pharmacologie , Agents neuromédiateurs/métabolisme
3.
Pak J Pharm Sci ; 37(4): 829-838, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39348648

RÉSUMÉ

Oxidative stress, stemming from heightened production of reactive oxygen species and free radicals, significantly contributes to the aging process. Apomorphine emerges as a pivotal medication for managing Alzheimer's, Parkinson's and other age-related conditions. This study aims to explore the memory-enhancing and neuroprotective properties of apomorphine, utilizing male Albino Wistar rats aged 4 and 24 months as subjects. Rats were intraperitoneally injected with apomorphine for 6 days. Decreased glutathione peroxidase, superoxide dismutase and catalase activities with increased lipid peroxidation were observed in the brain and plasma samples of aged rats, which were reversed upon apomorphine administration. Superoxide dismutase (SOD) and AChE activities were significantly decreased along with a decline in short-term- and long-term memory of aged rats, which was reverted by apomorphine. Furthermore, a notable reduction in biogenic amines and metabolite levels in the brains of aged rats was reversed in aged rats treated with apomorphine. The findings indicate a significant restoration of memory impairment and oxidative stress in aged rats by apomorphine. Overall, our data suggests that apomorphine, at a dosage of 1mg/kg, holds promise as a potential therapeutic intervention for dementia and associated disorders in elderly patients.


Sujet(s)
Apomorphine , Dysfonctionnement cognitif , Peroxydation lipidique , Troubles de la mémoire , Stress oxydatif , Rat Wistar , Animaux , Apomorphine/pharmacologie , Mâle , Troubles de la mémoire/traitement médicamenteux , Stress oxydatif/effets des médicaments et des substances chimiques , Dysfonctionnement cognitif/traitement médicamenteux , Dysfonctionnement cognitif/métabolisme , Peroxydation lipidique/effets des médicaments et des substances chimiques , Rats , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Superoxide dismutase/métabolisme , Vieillissement/effets des médicaments et des substances chimiques , Vieillissement/métabolisme , Acetylcholinesterase/métabolisme , Catalase/métabolisme , Neuroprotecteurs/pharmacologie , Glutathione peroxidase/métabolisme
4.
Turk J Med Sci ; 54(4): 866-875, 2024.
Article de Anglais | MEDLINE | ID: mdl-39295610

RÉSUMÉ

Background/aim: Diabetes mellitus, characterized by hyperglycemia, causes various complications, one of which is memory dysfunction. The frontal lobe is known to be responsible for impaired memory function due to hyperglycemia and is associated with oxidative stress-mediated neuronal cell apoptosis. Chlorogenic acid (CGA) is reported to have neuroprotective effects. However, its effect on the frontal lobe in diabetes mellitus (DM) rats is not widely known. This research aimed to elucidate the effect of CGA on the mRNA expressions of SOD1, SOD2, p53, and Bcl-2 in the frontal lobe of DM rats. Materials and methods: Thirty male Wistar rats (2-month-old, 150-200 gBW) were randomly divided into six groups: C (control), DM1.5 (1.5-month DM), DM2 (2-month DM), CGA12.5, CGA25 and CGA50 (DM+CGA 12.5, 25, and 50 mg/kgBW, respectively). A single dose of streptozotocin (60 mg/kgBW) was intraperitoneally injected. Intraperitoneal CGA injection was administered daily for DM1.5 rats for 14 days. Path length was measured in the Morris water maze (MWM) probe test. After termination, the frontal lobes were carefully harvested for RNA extraction. Reverse transcriptase PCR was performed to examine the mRNA expression of SOD1, SOD2, p53, and Bcl-2. Results: The DM2 group demonstrated significant shorter path length on the MWM probe test and significantly lower mRNA expression of SOD1 and Bcl-2, compared to the C group. After CGA administration, the CGA25 group showed a significantly shorter path length than the C group. The CGA12.5 and CGA25 groups had significantly higher mRNA expression of SOD1 than the DM1.5 group. Compared to the DM1.5 and DM2 groups, SOD2 mRNA expression of the administration of all three CGA doses increased markedly. Furthermore, Bcl-2 mRNA expression was significantly increased in the CGA12.5 and CGA50 groups, compared with the DM2 group. Conclusion: Chlorogenic acid might improve memory function through upregulation of frontal lobes' SOD1, SOD2, and Bcl-2 mRNA expression in DM rats.


Sujet(s)
Apoptose , Acide chlorogénique , Diabète expérimental , Lobe frontal , Troubles de la mémoire , Stress oxydatif , Rat Wistar , Animaux , Acide chlorogénique/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Mâle , Lobe frontal/effets des médicaments et des substances chimiques , Lobe frontal/métabolisme , Diabète expérimental/traitement médicamenteux , Diabète expérimental/complications , Rats , Apoptose/effets des médicaments et des substances chimiques , Troubles de la mémoire/traitement médicamenteux , Troubles de la mémoire/étiologie , Superoxide dismutase/métabolisme , Superoxide dismutase-1/génétique , Superoxide dismutase-1/métabolisme , Protéines proto-oncogènes c-bcl-2/métabolisme , Protéines proto-oncogènes c-bcl-2/génétique
5.
Int J Mol Sci ; 25(17)2024 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-39273620

RÉSUMÉ

The maintenance of proper brain function relies heavily on the balance of excitatory and inhibitory neural circuits, governed in part by synaptic adhesion molecules. Among these, MDGA1 (MAM domain-containing glycosylphosphatidylinositol anchor 1) acts as a suppressor of synapse formation by interfering with Neuroligin-mediated interactions, crucial for maintaining the excitatory-inhibitory (E/I) balance. Mdga1-/- mice exhibit selectively enhanced inhibitory synapse formation in their hippocampal pyramidal neurons, leading to impaired hippocampal long-term potentiation (LTP) and hippocampus-dependent learning and memory function; however, it has not been fully investigated yet if the reduction in MDGA1 protein levels would alter brain function. Here, we examined the behavioral and synaptic consequences of reduced MDGA1 protein levels in Mdga1+/- mice. As observed in Mdga1-/- mice, Mdga1+/- mice exhibited significant deficits in hippocampus-dependent learning and memory tasks, such as the Morris water maze and contextual fear-conditioning tests, along with a significant deficit in the long-term potentiation (LTP) in hippocampal Schaffer collateral CA1 synapses. The acute administration of D-cycloserine, a co-agonist of NMDAR (N-methyl-d-aspartate receptor), significantly ameliorated memory impairments and restored LTP deficits specifically in Mdga1+/- mice, while having no such effect on Mdga1-/- mice. These results highlight the critical role of MDGA1 in regulating inhibitory synapse formation and maintaining the E/I balance for proper cognitive function. These findings may also suggest potential therapeutic strategies targeting the E/I imbalance to alleviate cognitive deficits associated with neuropsychiatric disorders.


Sujet(s)
Cyclosérine , Haploinsuffisance , Hippocampe , Potentialisation à long terme , Troubles de la mémoire , Animaux , Potentialisation à long terme/effets des médicaments et des substances chimiques , Cyclosérine/pharmacologie , Souris , Troubles de la mémoire/traitement médicamenteux , Troubles de la mémoire/génétique , Troubles de la mémoire/métabolisme , Hippocampe/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Souris knockout , Mâle , Souris de lignée C57BL , Synapses/métabolisme , Synapses/effets des médicaments et des substances chimiques , Protéines liées au GPI/génétique , Protéines liées au GPI/métabolisme , Mémoire/effets des médicaments et des substances chimiques , Cellules pyramidales/métabolisme , Cellules pyramidales/effets des médicaments et des substances chimiques
6.
Nutrients ; 16(17)2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39275263

RÉSUMÉ

Recent studies have reported the benefits of food-derived peptides for memory dysfunction. Beyond the physiological effects of peptides, their bioavailability to the brain still remains unclear since the blood-brain barrier (BBB) strictly controls the transportation of compounds to the brain. Here, updated transportation studies on BBB transportable peptides are introduced and evaluated using in vitro BBB models, in situ perfusion, and in vivo mouse experiments. Additionally, the mechanisms of action of brain health peptides in relation to the pathogenesis of neurodegenerative diseases, particularly Alzheimer's disease, are discussed. This discussion follows a summary of bioactive peptides with neuroprotective effects that can improve cognitive decline through various mechanisms, including anti-inflammatory, antioxidative, anti-amyloid ß aggregation, and neurotransmitter regulation.


Sujet(s)
Maladie d'Alzheimer , Barrière hémato-encéphalique , Encéphale , Mémoire , Peptides , Animaux , Barrière hémato-encéphalique/métabolisme , Humains , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Mémoire/effets des médicaments et des substances chimiques , Mémoire/physiologie , Peptides/pharmacologie , Maladie d'Alzheimer/métabolisme , Transport biologique , Neuroprotecteurs/pharmacologie , Souris , Troubles de la mémoire/traitement médicamenteux
7.
J Alzheimers Dis ; 101(3): 797-811, 2024.
Article de Anglais | MEDLINE | ID: mdl-39240642

RÉSUMÉ

Background: Sigma-1 receptors are highly expressed in brain areas related to cognitive function and are a promising target for anti-amnesic treatments. We previously showed that activation of sigma-1 receptors by the selective agonist compound methyl(1 R,2 S/1 S,2 R)-2-[4-hydroxy-4-phenylpiperidin-1-yl)methyl]-1-(4-methylphenyl) cyclopropane carboxylate [(±)-PPCC] promotes a remarkable recovery in rat models of memory loss associated to cholinergic dysfunction. Objective: In this study, we sought to assess the role of (±)-PPCC on working memory deficits caused by noradrenergic depletion. Methods: Animals with a mild or severe working memory deficits associated to varying degrees of noradrenergic neuronal depletion were treated with the sigma-1 agonist just prior to the beginning of each behavioral testing session. Results: While (±)-PPCC alone at a dose of 1 mg/kg/day failed to affect working memory in lesioned animals, its association with the α2 adrenergic receptor agonist clonidine, completely blocked noradrenaline release, significantly improving rat performance. This effect, distinct from noradrenaline activity, is likely to result from a direct action of the (±)-PPCC compound onto sigma-1 receptors, as pre-treatment with the selective sigma-1 receptor antagonist BD-1047 reversed the improved working memory performance. Despite such clear functional effects, the treatment did not affect noradrenergic neuron survival or terminal fiber proliferation. Conclusions: Future studies are thus necessary to address the effects of long-lasting (±)-PPCC treatment, with or without clonidine, on cognitive abilities and Alzheimer's disease-like histopathology. Considering the already established involvement of sigma-1 receptors in endogenous cell plasticity mechanisms, their activation by selective agonist compounds holds promises as possibly positive contributor to disease-modifying events in neurodegenerative diseases.


Sujet(s)
Modèles animaux de maladie humaine , Troubles de la mémoire , Récepteur sigma , Sigma-1 Receptor , Animaux , Récepteur sigma/agonistes , Récepteur sigma/métabolisme , Troubles de la mémoire/traitement médicamenteux , Rats , Mâle , Rat Wistar , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques , Apprentissage du labyrinthe/physiologie , Pipéridines/pharmacologie , Cyclopropanes/pharmacologie , Cyclopropanes/usage thérapeutique
9.
Peptides ; 181: 171298, 2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39317295

RÉSUMÉ

Cocaine and amphetamine-regulated transcript (CART) mRNA and peptide are vastly expressed in both cortical and subcortical brain areas and are involved in critical cognitive functions. CART peptide (CARTp), described in reward-related brain structures, regulates drug-induced learning and memory, and its role appears specific to psychostimulants. However, many other drugs of abuse, such as alcohol, opiates, nicotine, and caffeine, have been shown to alter the expression levels of CART mRNA and peptides in brain structures directly or indirectly associated with learning and memory processes. However, the number of studies demonstrating the contribution of CARTp in learning and memory is still minimal. Notably, the exact cellular and molecular mechanisms underlying CARTp effects are still unknown. The discoveries that CARTp effects are mediated through a putative G-protein coupled receptor and activation of cellular signaling cascades via NMDA receptor-coupled ERK have enhanced our knowledge about the action of this neuropeptide and allowed us to comprehend better CARTp exact cellular/molecular mechanisms that could mediate drug-induced changes in learning and memory functions. Unfortunately, these efforts have been impeded by the lack of suitable and specific CARTp receptor antagonists. In this review, following a short introduction about CARTp, we report on current knowledge about CART's roles in learning and memory processes and its recently described role in memory-related neurological disorders. We will also discuss the importance of further investigating how CARTp interacts with its receptor(s) and other neurotransmitter systems to influence learning and memory functions. This topic is sure to intrigue and motivate further exploration in the field of neuroscience.


Sujet(s)
Apprentissage , Troubles de la mémoire , Mémoire , Protéines de tissu nerveux , Humains , Protéines de tissu nerveux/métabolisme , Mémoire/effets des médicaments et des substances chimiques , Mémoire/physiologie , Animaux , Apprentissage/effets des médicaments et des substances chimiques , Apprentissage/physiologie , Troubles de la mémoire/traitement médicamenteux , Troubles de la mémoire/métabolisme , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques
10.
Brain Res Bull ; 217: 111078, 2024 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-39270804

RÉSUMÉ

OBJECTIVE: The aim of this study is to elucidate the underlying mechanism through which glial cell line-derived neurotrophic factor (GDNF) improves cognitive deficits in adults resulting from neonatal surgical interventions. METHODS: Newborn Sprague-Dawley rats, regardless of gender, were randomly allocated into seven groups on postnatal day 7 as follows (n=15): (1) Control group (not subjected to anesthesia, surgery, or any pharmaceutical interventions); (2) GDNF group (received intracerebroventricular injection of GDNF); (3) Surgery group (underwent right carotid artery exposure under anesthesia with 3 % sevoflurane); (4) Surgery plus GDNF group; (5) Surgery plus GDNF and type II JAK inhibitor NVP-BBT594 (BBT594) group (administered intraperitoneal injection of BBT594); (6) BBT group; and (7) Surgery plus BBT group. Starting from postnatal day 33, all rats underwent Barnes maze and fear conditioning tests, followed by decapitation under sevoflurane anesthesia for subsequent analyses. The left hemibrains underwent Golgi staining, while the right hemibrains were used for hippocampal protein extraction to assess Protein kinase Mζ (PKMζ) and Kalirin expression through western blotting. RESULTS: GDNF demonstrated a mitigating effect on spatial learning and memory impairment, as well as context-related fear memory impairment, reductions in dendritic total lengths, and spinal density within the hippocampus induced by surgical intervention. Notably, all of these ameliorative effects of GDNF were reversed upon administration of the RET inhibitor BBT594. Additionally, GDNF alleviated the downregulation of protein expression of PKMζ and Kalirin in the hippocampus of rats subjected to surgery, subsequently reversed by BBT594. CONCLUSION: The effective impact of GDNF on learning and memory impairment caused by surgical intervention appears to be mediated through the RET pathway. Moreover, GDNF may exert its influence by upregulating the expression of PKMζ and Kalirin, consequently enhancing the development of dendrites and dendritic spines.


Sujet(s)
Animaux nouveau-nés , Facteur neurotrophique dérivé des cellules gliales , Rat Sprague-Dawley , Animaux , Facteur neurotrophique dérivé des cellules gliales/métabolisme , Rats , Mâle , Protéine kinase C/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/physiologie , Troubles de la mémoire/métabolisme , Troubles de la mémoire/traitement médicamenteux , Cognition/effets des médicaments et des substances chimiques , Cognition/physiologie , Femelle , Hippocampe/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Facteurs d'échange de nucléotides guanyliques/métabolisme , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques , Apprentissage du labyrinthe/physiologie , Récupération fonctionnelle/effets des médicaments et des substances chimiques , Récupération fonctionnelle/physiologie , Protéines proto-oncogènes c-ret
11.
J Med Chem ; 67(19): 17774-17784, 2024 Oct 10.
Article de Anglais | MEDLINE | ID: mdl-39321314

RÉSUMÉ

PDE11A4 is a target of interest for the treatment of age-related memory disorders. A previous report from our laboratories described an amide series of potent, selective PDE11A4 inhibitors that was metabolically unstable. Investigation of heterocyclic amide isosteres for the labile amide moiety revealed distinct structure-activity relationships and identified several compounds with potency comparable to the amide series. This manuscript describes the characterization of structure-activity and structure-property relationships in this set, leading to the identification of an orally bioavailable, brain-penetrant, selective and potent PDE11A4 inhibitor. Target engagement experiments demonstrated PDE11A4 inhibition in the hypothalamus of mice that was absent in PDE11A4 knock out animals.


Sujet(s)
Troubles de la mémoire , Animaux , Relation structure-activité , Souris , Troubles de la mémoire/traitement médicamenteux , Humains , Souris knockout , Mâle , Hypothalamus/métabolisme , Vieillissement
12.
BMC Neurosci ; 25(1): 45, 2024 Sep 27.
Article de Anglais | MEDLINE | ID: mdl-39333878

RÉSUMÉ

BACKGROUND: Exposure to chemical toxins, including insecticides, harms bodily organs like the brain. This study examined the neuroprotective of thymoquinone on the cypermethrin's harmful effects on the histoarchitecture of the dentate gyrus and motor deficit in the dentate gyrus. METHODS: Forty adult male rats (180-200 g) were randomly divided into 5 groups (n = 8 per group). Groups I, II, III, IV, and V received oral administration of 0.5 ml of phosphate-buffered saline, cypermethrin (20 mg/kg), thymoquinone (10 mg/kg), cypermethrin (20 mg/kg) + thymoquinone (5 mg/kg), and cypermethrin (20 mg/kg) + thymoquinone (10 mg/kg) for 14 days respectively. The novel object recognition test that assesses intermediate-term memory was done on days 14 and 21 of the experiment. At the end of these treatments, the animals were euthanized and taken for cytoarchitectural (hematoxylin and eosin; Cresyl violet) and immunohistochemical studies (Nuclear factor erythroid 2-related factor 2 (Nrf2), Parvalbumin, and B-cell lymphoma 2 (Bcl2). RESULT: The study shows that thymoquinone at 5 and 10 mg/kg improved Novelty preference and discrimination index. Thymoquinone enhanced Nissl body integrity, increased GABBAergic interneuron expression, nuclear factor erythroid 2-derived factor 2, and enhanced Bcl-2 expression in the dentate gyrus. It also improved the concentration of nuclear factor erythroid 2-derived factor 2, increased the activities of superoxide dismutase and glutathione, and decreased the concentration of malondialdehyde level against cypermethrin-induced neurotoxicity. CONCLUSION: thymoquinone could be a therapeutic agent against cypermethrin poisoning.


Sujet(s)
Benzoquinones , Gyrus denté , Neurones GABAergiques , Troubles de la mémoire , Facteur-2 apparenté à NF-E2 , Stress oxydatif , Pyréthrines , Transduction du signal , Animaux , Pyréthrines/toxicité , Mâle , Stress oxydatif/effets des médicaments et des substances chimiques , Benzoquinones/pharmacologie , Gyrus denté/effets des médicaments et des substances chimiques , Gyrus denté/métabolisme , Gyrus denté/anatomopathologie , Neurones GABAergiques/effets des médicaments et des substances chimiques , Neurones GABAergiques/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Troubles de la mémoire/induit chimiquement , Troubles de la mémoire/traitement médicamenteux , Rats , Facteur-2 apparenté à NF-E2/métabolisme , Insecticides/toxicité , Neuroprotecteurs/pharmacologie , Rat Wistar
13.
Alzheimers Res Ther ; 16(1): 200, 2024 Sep 07.
Article de Anglais | MEDLINE | ID: mdl-39244567

RÉSUMÉ

BACKGROUND: Synaptic dysfunction, characterized by synapse loss and structural alterations, emerges as a prominent correlate of cognitive decline in Alzheimer's disease (AD). Actin cytoskeleton, which serves as the structural backbone of synaptic architecture, is observed to be lost from synapses in AD. Actin cytoskeleton loss compromises synaptic integrity, affecting glutamatergic receptor levels, neurotransmission, and synaptic strength. Understanding these molecular changes is crucial for developing interventions targeting synaptic dysfunction, potentially mitigating cognitive decline in AD. METHODS: In this study, we investigated the synaptic actin interactome using mass spectrometry in a mouse model of AD, APP/PS1. Our objective was to explore how alterations in synaptic actin dynamics, particularly the interaction between PSD-95 and actin, contribute to synaptic and cognitive impairment in AD. To assess the impact of restoring F-actin levels on synaptic and cognitive functions in APP/PS1 mice, we administered F-actin stabilizing agent, jasplakinolide. Behavioral deficits in the mice were evaluated using the contextual fear conditioning paradigm. We utilized primary neuronal cultures to study the synaptic levels of AMPA and NMDA receptors and the dynamics of PSD-95 actin association. Furthermore, we analyzed postmortem brain tissue samples from subjects with no cognitive impairment (NCI), mild cognitive impairment (MCI), and Alzheimer's dementia (AD) to determine the association between PSD-95 and actin. RESULTS: We found a significant reduction in PSD-95-actin association in synaptosomes from middle-aged APP/PS1 mice compared to wild-type (WT) mice. Treatment with jasplakinolide, an actin stabilizer, reversed deficits in memory recall, restored PSD-95-actin association, and increased synaptic F-actin levels in APP/PS1 mice. Additionally, actin stabilization led to elevated synaptic levels of AMPA and NMDA receptors, enhanced dendritic spine density, suggesting improved neurotransmission and synaptic strength in primary cortical neurons from APP/PS1 mice. Furthermore, analysis of postmortem human tissue with NCI, MCI and AD subjects revealed disrupted PSD-95-actin interactions, underscoring the clinical relevance of our preclinical studies. CONCLUSION: Our study elucidates disrupted PSD-95 actin interactions across different models, highlighting potential therapeutic targets for AD. Stabilizing F-actin restores synaptic integrity and ameliorates cognitive deficits in APP/PS1 mice, suggesting that targeting synaptic actin regulation could be a promising therapeutic strategy to mitigate cognitive decline in AD.


Sujet(s)
Actines , Maladie d'Alzheimer , Souris transgéniques , Synapses , Animaux , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Actines/métabolisme , Souris , Synapses/métabolisme , Synapses/effets des médicaments et des substances chimiques , Humains , Mâle , Troubles de la mémoire/étiologie , Troubles de la mémoire/métabolisme , Troubles de la mémoire/traitement médicamenteux , Modèles animaux de maladie humaine , Femelle , Souris de lignée C57BL , Précurseur de la protéine bêta-amyloïde/génétique , Précurseur de la protéine bêta-amyloïde/métabolisme
14.
Brain Res ; 1844: 149196, 2024 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-39181223

RÉSUMÉ

This work was done to investigate the ameliorating impact of 4-methylumbilliferon (4-MU) on spatial learning and memory dysfunction and restraint stress (STR)-induced anxiety-like behaviors in male Wistar rats and the underlying mechanisms. Thirty-two animals were assigned into 4 cohorts: control, 4-MU, STR, and STR+4-MU. Animals were exposed to STR for 4 h per day for 14 consecutive days or kept in normal conditions (healthy animals without exposure to stress). 4-MU (25 mg/kg) was intraperitoneally administered once daily to STR rats before restraint stress for 14 consecutive days. The behavioral tests were performed through Morris water maze tests and elevated-plus maze to examine learning/memory function, and anxiety levels, respectively. The levels of the antioxidant defense biomarkers (GPX, SOD) and MDA as an oxidant molecule in the brain tissues were measured using commercial ELISA kits. Neuronal loss or density of neurons was evaluated using Nissl staining. STR exposure could cause significant alterations in the levels of the antioxidant defense biomarkers (MDA, GPX, and SOD) in the prefrontal cortex and hippocampus, induce anxiety, and impair spatial learning and memory function. Treatment with 4-MU markedly reduced anxiety levels and improved spatial learning and memory dysfunction via restoring the antioxidant defense biomarkers to normal values and reducing MDA levels. Moreover, more intact cells with normal morphologies were detected in STR-induced animals treated with 4-MU. 4-MU could attenuate the STR-induced anxiety-like behaviors and spatial learning and memory dysfunction by reducing oxidative damage and neuronal loss in the prefrontal cortex and hippocampus region. Taken together, our findings provide new insights regarding the potential therapeutic effects of 4-MU against neurobehavioral disorders induced by STR.


Sujet(s)
Anxiété , Mort cellulaire , Troubles de la mémoire , Neurones , Stress oxydatif , Rat Wistar , Animaux , Stress oxydatif/effets des médicaments et des substances chimiques , Mâle , Anxiété/traitement médicamenteux , Anxiété/métabolisme , Troubles de la mémoire/traitement médicamenteux , Troubles de la mémoire/métabolisme , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Rats , Mort cellulaire/effets des médicaments et des substances chimiques , Stress psychologique/traitement médicamenteux , Stress psychologique/métabolisme , Stress psychologique/complications , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Antioxydants/pharmacologie , Cortex préfrontal/effets des médicaments et des substances chimiques , Cortex préfrontal/métabolisme
15.
Behav Brain Res ; 474: 115175, 2024 10 02.
Article de Anglais | MEDLINE | ID: mdl-39098399

RÉSUMÉ

Stress-related disorders are becoming increasingly common and are often associated with cognitive impairments. Within this context, the endocannabinoid (ECB) system, particularly the type 1 cannabinoid (CB1) receptor, seems to play a decisive role in restoring body homeostasis. There is consistent evidence in the literature that disrupted CB1-mediated neurotransmission can ultimately contribute to stress-related diseases. Therefore, the present study aimed to evaluate the participation of CB1 receptors in the integrity of stress-induced peripheral and behavioral responses. For this purpose, male adult Wistar rats underwent physical restraint (1 h/day, for 7 days), followed by a single administration of rimonabant (CB1 receptor antagonist, 3 mg/Kg, intraperitonial) at the end of stress protocol. Animals were then subjected to evaluation of neuroendocrine responses, behavioral tests and quantification of Iba-1 (microglial) immunoreactivity in the parvocellular subdivisions of the paraventricular nucleus of the hypothalamus (PVN). No effects of restraint stress or rimonabant administration were detected on body mass variation. However, stress significantly increased adrenal relative mass and corticosterone secretion, and reduced thymus relative size. The stress effects on adrenal size and corticosterone plasma levels were absent in rimonabant-treated rats, but the thymus size was further reduced in the restraint-rimonabant group. Restraint stress also induced anhedonia, a depression-like behavior, and reduced object recognition index, indicating memory recovery impairment. Treatment with the CB1 antagonist significantly reversed stress-induced anhedonia and memory deficit. In the PVN, restraint stress reduced the number of Iba-1 positive cells in the medial parvocellular region of vehicle- but not rimonabant-treated animals. Taken together, these results indicate that the acute inhibition of the CB1-mediated endogenous pathway restores stress-induced depression-like behaviors and memory loss, suggesting a role for endocannabinoids in the neuro-immune-endocrine interplay at both peripheral and hypothalamic levels.


Sujet(s)
Anhédonie , Antagonistes des récepteurs de cannabinoïdes , Corticostérone , Troubles de la mémoire , Rat Wistar , Récepteur cannabinoïde de type CB1 , Contention physique , Rimonabant , Stress psychologique , Animaux , Rimonabant/pharmacologie , Mâle , Stress psychologique/métabolisme , Anhédonie/effets des médicaments et des substances chimiques , Anhédonie/physiologie , Rats , Troubles de la mémoire/traitement médicamenteux , Récepteur cannabinoïde de type CB1/métabolisme , Récepteur cannabinoïde de type CB1/antagonistes et inhibiteurs , Corticostérone/sang , Antagonistes des récepteurs de cannabinoïdes/pharmacologie , Noyau paraventriculaire de l'hypothalamus/métabolisme , Noyau paraventriculaire de l'hypothalamus/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Comportement animal/effets des médicaments et des substances chimiques , Pipéridines/pharmacologie , Pyrazoles/pharmacologie
16.
Behav Brain Res ; 472: 115174, 2024 08 24.
Article de Anglais | MEDLINE | ID: mdl-39098398

RÉSUMÉ

Sepsis-associated encephalopathy (SAE) is a common and severe clinical feature of sepsis; however, therapeutic approaches are limited because of the unclear pathogenesis. Adiponectin receptor agonist (AdipoRon) is a small-molecule agonist of the adiponectin receptor that exhibits anti-inflammatory and memory-improving effects in various diseases. In the present study, we established lipopolysaccharide (LPS)-induced mice models of SAE and found that Adiponectin receptor 1 (AdipoR1) was significantly decreased in the hippocampus. Administration of AdipoRon improves memory impairment, mitigates synaptic damage, and alleviates neuronal death. Furthermore, AdipoRon reduces the number of microglia. More importantly, AdipoRon promotes the phosphorylation of adenosine 5 '-monophosphate activated protein kinase (pAMPK). In conclusion, AdipoRon is protective against SAE-induced memory decline and brain injury in the SAE models via activating the hippocampal adenosine 5 '-monophosphate activated protein kinase (AMPK).


Sujet(s)
Modèles animaux de maladie humaine , Hippocampe , Troubles de la mémoire , Récepteurs à l'adiponectine , Animaux , Mâle , Souris , AMP-Activated Protein Kinases/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Lipopolysaccharides/pharmacologie , Troubles de la mémoire/traitement médicamenteux , Souris de lignée C57BL , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Pipéridines/pharmacologie , Récepteurs à l'adiponectine/agonistes , Récepteurs à l'adiponectine/métabolisme , Sepsie/traitement médicamenteux , Sepsie/complications , Sepsie/métabolisme , Encéphalopathie associée au sepsis/traitement médicamenteux , Encéphalopathie associée au sepsis/métabolisme
17.
Neurochem Int ; 179: 105834, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39142353

RÉSUMÉ

Alcohol exposure in adolescence is considered a major cause of cognitive impairments later in life including spatial learning and memory. Integrated stress response (ISR), a program of conservative translation and transcription, is crucial in synaptic plasticity and memory. Although previous studies have elucidated ISR in different brain areas involved in learning and memory disorders, the impact of ISR on learning and memory following adolescent alcohol exposure remains unclear. Here, we demonstrated that adolescent intermittent ethanol (AIE) exposure caused spatial learning and memory impairment, combined with neuronal damage in the medial prefrontal cortex (mPFC), nucleus accumbens (NAc) and hippocampus (HIP) in adult rats. Moreover, integrated stress response inhibitor (ISRIB) administration not only improved spatial learning and memory impairment and neuronal damage but also inhibited the endoplasmic reticulum stress (ER) and reversed changes in synaptic proteins. These findings suggested that ISRIB ameliorates AIE exposure-induced spatial learning and memory deficits by improving neural morphology and synaptic function through inhibiting ER stress signaling pathway in the mPFC, NAc and HIP in adulthood. Our findings may enhance comprehension of cognitive function and neuronal effects of adolescent ethanol exposure and ISRIB treatment may be an underlying potential option for addressing alcohol-induced learning and memory deficits.


Sujet(s)
Éthanol , Troubles de la mémoire , Rat Sprague-Dawley , Apprentissage spatial , Animaux , Mâle , Éthanol/toxicité , Éthanol/administration et posologie , Rats , Troubles de la mémoire/induit chimiquement , Troubles de la mémoire/traitement médicamenteux , Troubles de la mémoire/métabolisme , Apprentissage spatial/effets des médicaments et des substances chimiques , Cortex préfrontal/effets des médicaments et des substances chimiques , Cortex préfrontal/métabolisme , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques , Apprentissage du labyrinthe/physiologie
18.
Mol Biol Rep ; 51(1): 945, 2024 Aug 31.
Article de Anglais | MEDLINE | ID: mdl-39215798

RÉSUMÉ

Neurodegenerative diseases are characterized by progressive memory impairment and cognitive decline. This review aims to unravel the molecular mechanisms involved in the enhancement of memory function and mitigation of memory impairment through the activation of PPARγ agonists in neurodegenerative diseases. The findings suggest that PPARγ agonists modulate various molecular pathways involved in memory formation and maintenance. Activation of PPARγ enhances synaptic plasticity, promotes neuroprotection, suppresses neuroinflammation, attenuates oxidative stress, and regulates amyloid-beta metabolism. The comprehensive understanding of these molecular mechanisms would facilitate the development of novel therapeutic approaches targeting PPARγ to improve memory function and ultimately to alleviate the burden of neurodegenerative diseases. Further research, including clinical trials, is warranted to explore the efficacy, safety, and optimal use of specific PPARγ agonists as potential therapeutic agents in the treatment of memory impairments associated with neurodegenerative diseases.


Sujet(s)
Troubles de la mémoire , Maladies neurodégénératives , Stress oxydatif , Récepteur PPAR gamma , Humains , Récepteur PPAR gamma/agonistes , Récepteur PPAR gamma/métabolisme , Maladies neurodégénératives/traitement médicamenteux , Maladies neurodégénératives/métabolisme , Troubles de la mémoire/traitement médicamenteux , Troubles de la mémoire/métabolisme , Animaux , Stress oxydatif/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Plasticité neuronale/effets des médicaments et des substances chimiques , Mémoire/effets des médicaments et des substances chimiques , Peptides bêta-amyloïdes/métabolisme
19.
Behav Brain Res ; 474: 115180, 2024 10 02.
Article de Anglais | MEDLINE | ID: mdl-39111405

RÉSUMÉ

The present study aimed to assess the potential effect of vitamin B12 (Vit B12) on cognition impairment caused by nicotine (Nic) cessation in adolescent male rats. Adolescent male rats were categorized into two main groups as vehicle (normal saline, intraperitoneally), and Nic group in which received Nic (2 mg/kg) from 21 to 42 days of ages and then the Nic group were divided into three groups as withdrawal (the animals returned to regular diet without treatment), second and third groups received bupropion (20 mg/kg), and Vit B12 at three different doses including 0.5,1, and 1.5 mg/kg by oral gavage as treatments to attenuate Nic withdrawal symptoms. The last group including normal animals received the highest doses of Vit B12 just in the Nic abstinence period to compare the effect of that with vehicle. In MWM, Vit B12and bupropion increased the time spent in the target quadrant that is strongly associated with spatial memory as well as the more time spent with the NORT. Vit B12 and bupropion modulated both oxidant/antioxidant and inflammatory/anti-inflammatory balance, alongside inhibitory effect on AChE, and GFAP. However, BDNF and amyloid-B showed insignificant difference as compared to Vit B12 and bupropion. Considering the present results and similar related studies, Vit B12 can be introduced as a strong anti-oxidant, and anti-inflammatory agent by which probably improved memory impairment caused by Nic addiction accompanied by withdrawal. Further, other mechanisms including activity reduction of AChE, and GFAP should be considered; however, it needs further investigation and larger-scale evidences.


Sujet(s)
Facteur neurotrophique dérivé du cerveau , Protéine gliofibrillaire acide , Troubles de la mémoire , Nicotine , Syndrome de sevrage , Vitamine B12 , Animaux , Mâle , Rats , Acetylcholinesterase/métabolisme , Facteur neurotrophique dérivé du cerveau/métabolisme , Facteur neurotrophique dérivé du cerveau/effets des médicaments et des substances chimiques , Bupropion/pharmacologie , Bupropion/administration et posologie , Compléments alimentaires , Protéine gliofibrillaire acide/métabolisme , Inflammation/traitement médicamenteux , Troubles de la mémoire/traitement médicamenteux , Troubles de la mémoire/étiologie , Troubles de la mémoire/induit chimiquement , Nicotine/pharmacologie , Nicotine/administration et posologie , Stress oxydatif/effets des médicaments et des substances chimiques , Rat Wistar , Syndrome de sevrage/traitement médicamenteux , Vitamine B12/pharmacologie , Vitamine B12/administration et posologie
20.
Eur J Pharmacol ; 982: 176879, 2024 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-39128806

RÉSUMÉ

Alzheimer's disease (AD), the most common cause of dementia, leads to neurodegeneration and cognitive decline. We investigated the therapeutic effects of L-carnitine on cognitive performance and anxiety-like behavior in a rat model of AD induced by unilateral intracerebroventricular injection of ß-amyloid1-42 (Aß1-42). L-carnitine (100 mg/kg/day) was administered intraperitoneally for 28 consecutive days. Following this, the open-field test, novel object recognition test, elevated plus-maze test, Barnes maze test, and passive avoidance learning test were used to assess locomotor activity, recognition memory, anxiety-like behavior, spatial memory, and passive avoidance memory, respectively. Plasma and hippocampal oxidative stress markers, including total oxidant status (TOS) and total antioxidant capacity (TAC), were examined. In addition, histological investigations were performed in the dentate gyrus of the hippocampus using Congo red staining and hematoxylin and eosin staining. The injection of Aß1-42 resulted in cognitive deficits and increased anxiety-like behavior. These changes were associated with an imbalance of oxidants and antioxidants in plasma and the hippocampus. Also, neuronal death and Aß plaque accumulation were increased in the hippocampal dentate gyrus region. However, injection of L-carnitine improved recognition memory, spatial memory, and passive avoidance memory in AD rats. These findings provide evidence that L-carnitine may alleviate anxiety-like behavior and cognitive deficits induced by Aß1-42 through modulating oxidative-antioxidant status and preventing Aß plaque accumulation and neuronal death.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Anxiété , Carnitine , Modèles animaux de maladie humaine , Troubles de la mémoire , Stress oxydatif , Rat Wistar , Animaux , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/psychologie , Maladie d'Alzheimer/métabolisme , Peptides bêta-amyloïdes/métabolisme , Anxiété/traitement médicamenteux , Mâle , Troubles de la mémoire/traitement médicamenteux , Troubles de la mémoire/prévention et contrôle , Rats , Carnitine/pharmacologie , Carnitine/usage thérapeutique , Stress oxydatif/effets des médicaments et des substances chimiques , Comportement animal/effets des médicaments et des substances chimiques , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Fragments peptidiques , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Apprentissage par évitement/effets des médicaments et des substances chimiques , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques , Antioxydants/pharmacologie , Antioxydants/usage thérapeutique , Mémoire spatiale/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE