Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 643
Filtrer
1.
Eur J Neurosci ; 60(5): 4707-4722, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39119857

RÉSUMÉ

Disputes about the scientific validity of the amyloid-ß hypothesis of Alzheimer's disease have been held since the early 1990s, with little constructive progress made between opposing sides despite recent therapeutic progress. Here, I argue that philosophy of science can improve the chance of constructive debate by giving researchers technical language to describe and assess scientific progress. To do so, I interpret the amyloid hypothesis using a modified version of the research programme concept from philosopher of science Imre Lakatos. I first outline the amyloid-ß hypothesis and study critiques of its central place in Alzheimer's research. Then, I draw on the complexity of amyloid-ß and Alzheimer's research to discuss the limits of using concepts from popular philosophers of science Karl Popper or Thomas Kuhn, before finally arguing that an adaptation of the research programme concept can foster constructive debates about the science of Alzheimer's and within it. I will argue that the amyloid-ß hypothesis has contributed to significant progress in the Alzheimer's field based on what Lakatos called the "positive heuristic" (motivating the programme to test its predictions) and the "negative heuristic" (protecting the programme from refutation). I consider the amyloid research agenda to be progressive despite the fact that its claims about disease aetiology could be wrong.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Maladie d'Alzheimer/métabolisme , Humains , Peptides bêta-amyloïdes/métabolisme , Philosophie , Animaux
2.
J Phys Chem B ; 128(29): 7022-7032, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39016210

RÉSUMÉ

The interaction between iron and amyloid-beta (Aß) peptides has received significant attention in Alzheimer's disease (AD) research due to its potential implications in developing this pathology. However, the coordination preferences of iron and Aß1-42 have not been thoroughly investigated or remain unknown. This study employs a computational protocol that combines homology modeling techniques with quantum mechanics (DTF-xTB) calculations to build and evaluate several 3D models of Fe2+/3+-Aß1-42. Our results reveal well-defined complexes for both the metal and peptide moieties, and we discuss the molecular interactions stabilizing these complexes by elucidating the coordinating environments and binding preferences. These proposed models offer valuable insights into the role of iron in Alzheimer's disease (AD) pathology.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Fragments peptidiques , Peptides bêta-amyloïdes/composition chimique , Peptides bêta-amyloïdes/métabolisme , Maladie d'Alzheimer/métabolisme , Fragments peptidiques/composition chimique , Fragments peptidiques/métabolisme , Humains , Théorie quantique , Modèles moléculaires , Fer/composition chimique , Fer/métabolisme , Composés du fer III/composition chimique , Composés du fer II/composition chimique
3.
Ageing Res Rev ; 100: 102439, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39074563

RÉSUMÉ

Alzheimer's disease (AD) constitutes a major public-health issue of our time. Regrettably, despite our considerable understanding of the pathophysiological aspects of this disease, current interventions lead to poor outcomes. Furthermore, experimentally promising compounds have continuously failed when translated to clinical trials. Along with increased population ageing, Type 2 Diabetes Mellitus (T2DM) has become an extremely common condition, mainly due to unbalanced dietary habits. Substantial epidemiological evidence correlates T2DM with cognitive impairment as well. Considering that brain insulin resistance, mitochondrial dysfunction, oxidative stress, and amyloidogenesis are common phenomena, further approaching the common features among these pathological conditions. Metformin constitutes the first-choice drug to preclude insulin resistance in T2DM clinical management. Experimental evidence suggests that its functions might include neuroprotective effects, in addition to its hypoglycemic activity. This review aims to summarize and discuss current knowledge of experimental data on metformin on this path towards translational medicine. Finally, we discuss the controversial data of responses to metformin in vitro, and in vivo, animal models and human studies.


Sujet(s)
Maladie d'Alzheimer , Diabète de type 2 , Hypoglycémiants , Metformine , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Metformine/usage thérapeutique , Metformine/pharmacologie , Humains , Animaux , Hypoglycémiants/usage thérapeutique , Hypoglycémiants/pharmacologie , Diabète de type 2/traitement médicamenteux , Diabète de type 2/métabolisme , 53784/méthodes , Neuroprotecteurs/usage thérapeutique , Neuroprotecteurs/pharmacologie , Insulinorésistance/physiologie
4.
Neurochem Res ; 49(10): 2785-2802, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-38985243

RÉSUMÉ

To contribute to research on female models of Alzheimer's disease (AD), our aim was to study the effect of intracerebroventricular (ICV) injection of streptozotocin (STZ) in female rats, and to evaluate a potential neuroprotective action of ovarian steroids against STZ. Female rats were either ovariectomized (OVX) or kept with ovaries (Sham) two weeks before ICV injections. Animals were injected with either vehicle (artificial cerebrospinal fluid, aCSF) or STZ (3 mg/kg) and separated into four experimental groups: Sham + aCSF, Sham + STZ, OVX + aCSF and OVX + STZ. Nineteen days post-injection, we assessed different behavioral aspects: burying, anxiety and exploration, object recognition memory, spatial memory, and depressive-like behavior. Immunohistochemistry and Immunoblot analyses were performed in the hippocampus to examine changes in AD-related proteins and neuronal and microglial populations. STZ affected burying and exploratory behavior depending on ovarian status, and impaired recognition but not spatial memory. STZ and ovariectomy increased depressive-like behavior. Interestingly, STZ did not alter the expression of ß-amyloid peptide or Tau phosphorylated forms. STZ affected the neuronal population from the Dentate Gyrus, where immature neurons were more vulnerable to STZ in OVX rats. Regarding microglia, STZ increased reactive cells, and the OVX + STZ group showed an increase in the total cell number. In sum, STZ partially affected female rats, compared to what was previously reported for males. Although AD is more frequent in women, reports about the effect of ICV-STZ in female rats are scarce. Our work highlights the need to deepen into the effects of STZ in the female brain and study possible sex differences.


Sujet(s)
Maladie d'Alzheimer , Ovariectomie , Streptozocine , Animaux , Femelle , Maladie d'Alzheimer/induit chimiquement , Maladie d'Alzheimer/métabolisme , Rats , Injections ventriculaires , Rat Wistar , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Ovaire/effets des médicaments et des substances chimiques , Ovaire/métabolisme , Dépression/induit chimiquement , Dépression/métabolisme , Peptides bêta-amyloïdes/métabolisme , Mémoire spatiale/effets des médicaments et des substances chimiques
5.
Basic Clin Pharmacol Toxicol ; 135(3): 237-249, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39020526

RÉSUMÉ

Alzheimer's disease is characterized by progressive cognitive decline, and behavioural and psychological symptoms of dementia are common. The APOE ε4 allele, a genetic risk factor, significantly increases susceptibility to the disease. Despite efforts to effectively treat the disease, only seven drugs are approved for its treatment, and only two of these prevent its progression. This highlights the need to identify new pharmacological options. This review focuses on mimetic peptides, small molecule correctors and HAE-4 antibodies that target ApoE. These drugs reduce ß-amyloid-induced neurodegeneration in preclinical models. In addition, loop diuretics such as bumetanide and furosemide show the potential to reduce the prevalence of Alzheimer's disease in humans, and antidepressants such as imipramine improve cognitive function in individuals diagnosed with Alzheimer's disease. Consistent with this, both classes of drugs have been shown to exert neuroprotective effects by inhibiting ApoE4-catalysed Aß aggregation in preclinical models. Moreover, peroxisome proliferator-activated receptor ligands, particularly pioglitazone and rosiglitazone, reduce ApoE4-induced neurodegeneration in animal models. However, they do not prevent the cognitive decline in APOE ε4 allele carriers. Finally, ApoE4 impairs the integrity of the blood-brain barrier and haemostasis. On this basis, ApoE4 modulation is a promising avenue for the treatment of late-onset Alzheimer's disease.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Apolipoprotéine E4 , Encéphale , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Humains , Apolipoprotéine E4/métabolisme , Apolipoprotéine E4/génétique , Animaux , Peptides bêta-amyloïdes/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Encéphale/anatomopathologie , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Agrégation pathologique de protéines/traitement médicamenteux , Agrégation pathologique de protéines/métabolisme , Modèles animaux de maladie humaine
6.
ACS Chem Neurosci ; 15(16): 2982-2994, 2024 Aug 21.
Article de Anglais | MEDLINE | ID: mdl-39007352

RÉSUMÉ

Alzheimer's disease (AD) is a complex neurodegenerative process, also considered a metabolic condition due to alterations in glucose metabolism and insulin signaling pathways in the brain, which share similarities with diabetes. This study aimed to investigate the therapeutic effects of benfotiamine (BFT), a vitamin B1 analog, in the early stages of the neurodegenerative process in a sporadic model of Alzheimer's-like disease induced by intracerebroventricular injection of streptozotocin (STZ). Supplementation with 150 mg/kg of BFT for 7 days reversed the cognitive impairment in short- and long-term memories caused by STZ in rodents. We attribute these effects to BFT's ability to modulate glucose transporters type 1 and 3 (GLUT1 and GLUT3) in the hippocampus, inhibit GSK3 activity in the hippocampus, and modulate the insulin signaling in the hippocampus and entorhinal cortex, as well as reduce the activation of apoptotic pathways (BAX) in the hippocampus. Therefore, BFT emerges as a promising and accessible intervention in the initial treatment of conditions similar to AD.


Sujet(s)
Maladie d'Alzheimer , Modèles animaux de maladie humaine , Hippocampe , Insuline , Transduction du signal , Streptozocine , Thiamine , Animaux , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Insuline/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Thiamine/pharmacologie , Thiamine/analogues et dérivés , Thiamine/usage thérapeutique , Rats , Cognition/effets des médicaments et des substances chimiques , Rat Wistar , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques
7.
Neurochem Res ; 49(9): 2535-2555, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38888830

RÉSUMÉ

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the accumulation of amyloid-ß, leading to N-methyl-D-aspartate (NMDA) receptor-dependent synaptic depression, spine elimination, and memory deficits. Glycine transporter type 1 (GlyT1) modulates glutamatergic neurotransmission via NMDA receptors (NMDAR), presenting a potential alternative therapeutic approach for AD. This study investigates the neuroprotective potential of GlyT1 inhibition in an amyloid-ß-induced AD mouse model. C57BL/6 mice were treated with N-[3-([1,1-Biphenyl]-4-yloxy)-3-(4-fluorophenyl)propyl]-N-methylglycine (NFPS), a GlyT1 inhibitor, 24 h prior to intrahippocampal injection of amyloid-ß. NFPS pretreatment prevented amyloid-ß-induced cognitive deficits in short-term and long-term memory, evidenced by novel object recognition and spatial memory tasks. Moreover, NFPS pretreatment curbed microglial activation, astrocytic reactivity, and subsequent neuronal damage from amyloid-ß injection. An extensive label-free quantitative UPLC-MSE proteomic analysis was performed on the hippocampi of mice treated with NFPS. In proteomics, KEGG enrichment analysis revealed increased in dopaminergic synapse, purine-containing compound biosynthetic process and long-term potentiation, and a reduction in Glucose catabolic process and glycolytic process pathways. The western blot analysis confirmed that NFPS treatment elevated BDNF levels, correlating with enhanced TRKB phosphorylation and mTOR activation. Moreover, NFPS treatment reduced the GluN2B expression after 6 h, which was associated with an increase on CaMKIV and CREB phosphorylation. Collectively, these findings demonstrate that GlyT1 inhibition by NFPS activates diverse neuroprotective pathways, enhancing long-term potentiation signaling and countering amyloid-ß-induced hippocampal damage.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Transporteurs de la glycine , Hippocampe , Souris de lignée C57BL , Neuroprotecteurs , Animaux , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/induit chimiquement , Maladie d'Alzheimer/prévention et contrôle , Peptides bêta-amyloïdes/métabolisme , Mâle , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Souris , Hippocampe/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Transporteurs de la glycine/antagonistes et inhibiteurs , Transporteurs de la glycine/métabolisme , Modèles animaux de maladie humaine , Sarcosine/analogues et dérivés , Sarcosine/pharmacologie , Sarcosine/usage thérapeutique , Neuroprotection/effets des médicaments et des substances chimiques , Neuroprotection/physiologie
8.
Alzheimers Dement ; 20(8): 5398-5410, 2024 08.
Article de Anglais | MEDLINE | ID: mdl-38934107

RÉSUMÉ

INTRODUCTION: Impaired brain protein synthesis, synaptic plasticity, and memory are major hallmarks of Alzheimer's disease (AD). The ketamine metabolite (2R,6R)-hydroxynorketamine (HNK) has been shown to modulate protein synthesis, but its effects on memory in AD models remain elusive. METHODS: We investigated the effects of HNK on hippocampal protein synthesis, long-term potentiation (LTP), and memory in AD mouse models. RESULTS: HNK activated extracellular signal-regulated kinase 1/2 (ERK1/2), mechanistic target of rapamycin (mTOR), and p70S6 kinase 1 (S6K1)/ribosomal protein S6 signaling pathways. Treatment with HNK rescued hippocampal LTP and memory deficits in amyloid-ß oligomers (AßO)-infused mice in an ERK1/2-dependent manner. Treatment with HNK further corrected aberrant transcription, LTP and memory in aged APP/PS1 mice. DISCUSSION: Our findings demonstrate that HNK induces signaling and transcriptional responses that correct synaptic and memory deficits in AD mice. These results raise the prospect that HNK could serve as a therapeutic approach in AD. HIGHLIGHTS: The ketamine metabolite HNK activates hippocampal ERK/mTOR/S6 signaling pathways. HNK corrects hippocampal synaptic and memory defects in two mouse models of AD. Rescue of synaptic and memory impairments by HNK depends on ERK signaling. HNK corrects aberrant transcriptional signatures in APP/PS1 mice.


Sujet(s)
Maladie d'Alzheimer , Modèles animaux de maladie humaine , Hippocampe , Kétamine , Souris transgéniques , Plasticité neuronale , Animaux , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Kétamine/analogues et dérivés , Kétamine/pharmacologie , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Plasticité neuronale/effets des médicaments et des substances chimiques , Souris , Potentialisation à long terme/effets des médicaments et des substances chimiques , Peptides bêta-amyloïdes/métabolisme , Biosynthèse des protéines/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme , ARN messager/métabolisme , Mémoire/effets des médicaments et des substances chimiques , Mâle , Troubles de la mémoire/traitement médicamenteux , Souris de lignée C57BL , Précurseur de la protéine bêta-amyloïde/génétique , Précurseur de la protéine bêta-amyloïde/métabolisme , Préséniline-1/génétique , Humains
9.
Front Immunol ; 15: 1401800, 2024.
Article de Anglais | MEDLINE | ID: mdl-38933275

RÉSUMÉ

Air pollution is an urgent concern linked to numerous health problems in low- and middle-income countries, where 92% of air pollution-related deaths occur. Particulate matter 2.5 (PM2.5) is the most harmful component of air pollutants, increasing inflammation and changing gut microbiota, favoring obesity, type 2 diabetes, and Alzheimer's Disease (AD). PM2.5 contains lipopolysaccharides (LPS), which can activate the Toll-like receptor 4 (TLR4) signaling pathway. This pathway can lead to the release of pro-inflammatory markers, including interleukins, and suppressor of cytokine signaling-3 (SOCS3), which inhibits leptin action, a hormone that keeps the energy homeostasis. Leptin plays a role in preventing amyloid plaque deposition and hyperphosphorylation of tau-protein (p-tau), mechanisms involved in the neurodegeneration in AD. Approximately 50 million people worldwide are affected by dementia, with a significant proportion living in low-and middle-income countries. This number is expected to triple by 2050. This mini-review focuses on the potential impact of PM2.5 exposure on the TLR4 signaling pathway, its contribution to leptin resistance, and dysbiosis that exacerbates the link between obesity and AD.


Sujet(s)
Pollution de l'air , Maladie d'Alzheimer , Inflammation , Leptine , Obésité , Matière particulaire , Animaux , Humains , Polluants atmosphériques/effets indésirables , Pollution de l'air/effets indésirables , Maladie d'Alzheimer/étiologie , Maladie d'Alzheimer/métabolisme , Inflammation/métabolisme , Inflammation/étiologie , Leptine/métabolisme , Obésité/métabolisme , Obésité/étiologie , Matière particulaire/effets indésirables , Transduction du signal , Récepteur de type Toll-4/métabolisme
10.
J Biol Inorg Chem ; 29(4): 407-425, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38811408

RÉSUMÉ

The influence of metal ions on the structure of amyloid- ß (Aß) protofibril models was studied through molecular dynamics to explore the molecular mechanisms underlying metal-induced Aß aggregation relevant in Alzheimer's disease (AD). The models included 36-, 48-, and 188-mers of the Aß42 sequence and two disease-modifying variants. Primary structural effects were observed at the N-terminal domain, as it became susceptible to the presence of cations. Specially when ß-sheets predominate, this motif orients N-terminal acidic residues toward one single face of the ß-sheet, resulting in the formation of an acidic region that attracts cations from the media and promotes the folding of the N-terminal region, with implications in amyloid aggregation. The molecular phenotype of the protofibril models based on Aß variants shows that the AD-causative D7N mutation promotes the formation of N-terminal ß-sheets and accumulates more Zn2+, in contrast to the non-amyloidogenic rodent sequence that hinders the ß-sheets and is more selective for Na+ over Zn2+ cations. It is proposed that forming an acidic ß-sheet domain and accumulating cations is a plausible molecular mechanism connecting the elevated affinity and concentration of metals in Aß fibrils to their high content of ß-sheet structure at the N-terminal sequence.


Sujet(s)
Peptides bêta-amyloïdes , Simulation de dynamique moléculaire , Peptides bêta-amyloïdes/composition chimique , Peptides bêta-amyloïdes/métabolisme , Peptides bêta-amyloïdes/génétique , Structure en brin bêta , Humains , Zinc/métabolisme , Zinc/composition chimique , Maladie d'Alzheimer/métabolisme , Fragments peptidiques/composition chimique , Fragments peptidiques/métabolisme , Fragments peptidiques/génétique , Métaux/métabolisme , Métaux/composition chimique
11.
J Alzheimers Dis ; 99(2): 639-656, 2024.
Article de Anglais | MEDLINE | ID: mdl-38728184

RÉSUMÉ

Background: Familial Alzheimer's disease (FAD) presenilin 1 E280A (PSEN 1 E280A) is characterized by functional impairment and the death of cholinergic neurons as a consequence of amyloid-ß (Aß) accumulation and abnormal phosphorylation of the tau protein. Currently, there are no available therapies that can cure FAD. Therefore, new therapies are urgently needed for treating this disease. Objective: To assess the effect of sildenafil (SIL) on cholinergic-like neurons (ChLNs) harboring the PSEN 1 E280A mutation. Methods: Wild-type (WT) and PSEN 1 E280A ChLNs were cultured in the presence of SIL (25µM) for 24 h. Afterward, proteinopathy, cell signaling, and apoptosis markers were evaluated via flow cytometry and fluorescence microscopy. Results: We found that SIL was innocuous toward WT PSEN 1 ChLNs but reduced the accumulation of intracellular Aß fragments by 87%, decreased the non-physiological phosphorylation of the protein tau at residue Ser202/Thr205 by 35%, reduced the phosphorylation of the proapoptotic transcription factor c-JUN at residue Ser63/Ser73 by 63%, decreased oxidized DJ-1 at Cys106-SO3 by 32%, and downregulated transcription factor TP53 (tumor protein p53), BH-3-only protein PUMA (p53 upregulated modulator of apoptosis), and cleaved caspase 3 (CC3) expression by 20%, 32%, and 22%, respectively, compared with untreated mutant ChLNs. Interestingly, SIL also ameliorated the dysregulation of acetylcholine-induced calcium ion (Ca2+) influx in PSEN 1 E280A ChLNs. Conclusions: Although SIL showed no antioxidant capacity in the oxygen radical absorbance capacity and ferric ion reducing antioxidant power assays, it might function as an anti-amyloid and antiapoptotic agent and functional neuronal enhancer in PSEN 1 E280A ChLNs. Therefore, the SIL has therapeutic potential for treating FAD.


Sujet(s)
Maladie d'Alzheimer , Neurones cholinergiques , Mutation , Préséniline-1 , Citrate de sildénafil , Préséniline-1/génétique , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/métabolisme , Neurones cholinergiques/effets des médicaments et des substances chimiques , Neurones cholinergiques/métabolisme , Neurones cholinergiques/anatomopathologie , Mutation/génétique , Animaux , Citrate de sildénafil/pharmacologie , Peptides bêta-amyloïdes/métabolisme , Humains , Cellules cultivées , Souris , Protéines tau/métabolisme , Protéines tau/génétique , Phosphorylation/effets des médicaments et des substances chimiques , Phénotype
12.
Prog Neurobiol ; 237: 102616, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38723884

RÉSUMÉ

Alterations in cognitive and non-cognitive cerebral functions characterize Alzheimer's disease (AD). Cortical and hippocampal impairments related to extracellular accumulation of Aß in AD animal models have been extensively investigated. However, recent reports have also implicated intracellular Aß in limbic regions, such as the nucleus accumbens (nAc). Accumbal neurons express high levels of inhibitory glycine receptors (GlyRs) that are allosterically modulated by ethanol and have a role in controlling its intake. In the present study, we investigated how GlyRs in the 2xTg mice (AD model) affect nAc functions and ethanol intake behavior. Using transgenic and control aged-matched litter mates, we found that the GlyRα2 subunit was significantly decreased in AD mice (6-month-old). We also examined intracellular calcium dynamics using the fluorescent calcium protein reporter GCaMP in slice photometry. We also found that the calcium signal mediated by GlyRs, but not GABAAR, was also reduced in AD neurons. Additionally, ethanol potentiation was significantly decreased in accumbal neurons in the AD mice. Finally, we performed drinking in the dark (DID) experiments and found that 2xTg mice consumed less ethanol on the last day of DID, in agreement with a lower blood ethanol concentration. 2xTg mice also showed lower sucrose consumption, indicating that overall food reward was altered. In conclusion, the data support the role of GlyRs in nAc neuron excitability and a decreased glycinergic activity in the 2xTg mice that might lead to impairment in reward processing at an early stage of the disease.


Sujet(s)
Maladie d'Alzheimer , Noyau accumbens , Récepteur de la glycine , Souris , Maladie d'Alzheimer/métabolisme , Noyau accumbens/métabolisme , Récepteur de la glycine/métabolisme , Souris de lignée C57BL , Éthanol , Souris transgéniques , Calcium/métabolisme , Récompense , Saccharose/métabolisme , Activité motrice , Anxiété , Neurones/métabolisme
13.
Neurochem Res ; 49(7): 1851-1862, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38733521

RÉSUMÉ

Alzheimer's disease (AD) is an age-dependent neurodegenerative disease that is typically sporadic and has a high social and economic cost. We utilized the intracerebroventricular administration of streptozotocin (STZ), an established preclinical model for sporadic AD, to investigate hippocampal astroglial changes during the first 4 weeks post-STZ, a period during which amyloid deposition has yet to occur. Astroglial proteins aquaporin 4 (AQP-4) and connexin-43 (Cx-43) were evaluated, as well as claudins, which are tight junction (TJ) proteins in brain barriers, to try to identify changes in the glymphatic system and brain barrier during the pre-amyloid phase. Glial commitment, glucose hypometabolism and cognitive impairment were characterized during this phase. Astroglial involvement was confirmed by an increase in glial fibrillary acidic protein (GFAP); concurrent proteolysis was also observed, possibly mediated by calpain. Levels of AQP-4 and Cx-43 were elevated in the fourth week post-STZ, possibly accelerating the clearance of extracellular proteins, since these proteins actively participate in the glymphatic system. Moreover, although we did not see a functional disruption of the blood-brain barrier (BBB) at this time, claudin 5 (present in the TJ of the BBB) and claudin 2 (present in the TJ of the blood-cerebrospinal fluid barrier) were reduced. Taken together, data support a role for astrocytes in STZ brain damage, and suggest that astroglial dysfunction accompanies or precedes neuronal damage in AD.


Sujet(s)
Maladie d'Alzheimer , Aquaporine-4 , Astrocytes , Streptozocine , Astrocytes/métabolisme , Animaux , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Mâle , Aquaporine-4/métabolisme , Connexine 43/métabolisme , Barrière hémato-encéphalique/métabolisme , Eau/métabolisme , Hippocampe/métabolisme , Rat Wistar , Rats , Modèles animaux de maladie humaine
14.
J Mater Chem B ; 12(21): 5085-5097, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38713059

RÉSUMÉ

Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by cognitive impairment associated with the accumulation of beta-amyloid protein (Aß). Aß activates glial cells in the brain, increasing the secretion of proinflammatory cytokines, which leads to neuroinflammation and neuronal death. Currently, there are no effective treatments that cure or stop its progression; therefore, AD is considered a global health priority. The main limitations are the low drug bioavailability and impermeability of the blood-brain barrier (BBB). Fortunately, nanomedicine has emerged as a promising field for the development of new nanosystems for the controlled and targeted delivery of drugs to the brain. Therefore, in this work, lipid-polymer hybrid nanoparticles (LPHNPs) conjugated with transferrin (Tf) to facilitate crossing the BBB and loaded with N-acetylcysteine (NAC) for its anti-inflammatory effect were synthesized, and their physicochemical characterization was carried out. Subsequently, an in vitro model involving human astrocytes derived from induced pluripotent stem cells (iPSC) from an AD-diagnosed patient was developed, which was brought to a reactive state by stimulation with lipopolysaccharides (LPSs). The cell culture was treated with either Tf-conjugated LPHNPs loaded with NAC (NAC-Tf-LPHNPs) at 0.25 mg mL-1, or free NAC at 5 mM. The results showed that NAC-Tf-LPHNPs favorably modulated the expression of proinflammatory genes such as interleukin-1ß (IL-1ß), amyloid precursor protein (APP) and glial fibrillary acidic protein (GFAP). In addition, they reduced the secretion of the proinflammatory cytokines interleukin 6 (IL-6), IL-1ß and interferon-gamma (INF-γ). Results for both cases were compared to the group of cells that did not receive any treatment. In contrast, free NAC only had this effect on the expression of IL-1ß and the secretion of the cytokines IL-6 and INF-γ. These results indicate the potential of NAC-Tf-LPHNPs for AD treatment.


Sujet(s)
Acétylcystéine , Maladie d'Alzheimer , Astrocytes , Cellules souches pluripotentes induites , Nanoparticules , Humains , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Nanoparticules/composition chimique , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques , Acétylcystéine/composition chimique , Acétylcystéine/pharmacologie , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Polymères/composition chimique , Polymères/pharmacologie , Lipides/composition chimique , Marqueurs biologiques/métabolisme , Taille de particule , Maladies neuro-inflammatoires/traitement médicamenteux
15.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38732141

RÉSUMÉ

Familial Alzheimer's disease (FAD) is a complex and multifactorial neurodegenerative disorder for which no curative therapies are yet available. Indeed, no single medication or intervention has proven fully effective thus far. Therefore, the combination of multitarget agents has been appealing as a potential therapeutic approach against FAD. Here, we investigated the potential of combining tramiprosate (TM), curcumin (CU), and the JNK inhibitor SP600125 (SP) as a treatment for FAD. The study analyzed the individual and combined effects of these two natural agents and this pharmacological inhibitor on the accumulation of intracellular amyloid beta iAß; hyperphosphorylated protein TAU at Ser202/Thr205; mitochondrial membrane potential (ΔΨm); generation of reactive oxygen species (ROS); oxidized protein DJ-1; proapoptosis proteins p-c-JUN at Ser63/Ser73, TP53, and cleaved caspase 3 (CC3); and deficiency in acetylcholine (ACh)-induced transient Ca2+ influx response in cholinergic-like neurons (ChLNs) bearing the mutation I416T in presenilin 1 (PSEN1 I416T). We found that single doses of TM (50 µM), CU (10 µM), or SP (1 µM) were efficient at reducing some, but not all, pathological markers in PSEN 1 I416T ChLNs, whereas a combination of TM, CU, and SP at a high (50, 10, 1 µM) concentration was efficient in diminishing the iAß, p-TAU Ser202/Thr205, DJ-1Cys106-SO3, and CC3 markers by -50%, -75%, -86%, and -100%, respectively, in PSEN1 I417T ChLNs. Although combinations at middle (10, 2, 0.2) and low (5, 1, 0.1) concentrations significantly diminished p-TAU Ser202/Thr205, DJ-1Cys106-SO3, and CC3 by -69% and -38%, -100% and -62%, -100% and -62%, respectively, these combinations did not alter the iAß compared to untreated mutant ChLNs. Moreover, a combination of reagents at H concentration was able to restore the dysfunctional ACh-induced Ca2+ influx response in PSEN 1 I416T. Our data suggest that the use of multitarget agents in combination with anti-amyloid (TM, CU), antioxidant (e.g., CU), and antiapoptotic (TM, CU, SP) actions might be beneficial for reducing iAß-induced ChLN damage in FAD.


Sujet(s)
Maladie d'Alzheimer , Anthracènes , Curcumine , Préséniline-1 , Taurine/analogues et dérivés , Curcumine/pharmacologie , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Préséniline-1/génétique , Préséniline-1/métabolisme , Anthracènes/pharmacologie , Animaux , Espèces réactives de l'oxygène/métabolisme , Souris , Peptides bêta-amyloïdes/métabolisme , Humains , Protéines tau/métabolisme , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Neurones/anatomopathologie , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques
16.
Sci Rep ; 14(1): 11268, 2024 05 17.
Article de Anglais | MEDLINE | ID: mdl-38760448

RÉSUMÉ

We aimed to study atrophy and glucose metabolism of the cholinergic basal forebrain in non-demented mutation carriers for autosomal dominant Alzheimer's disease (ADAD). We determined the level of evidence for or against atrophy and impaired metabolism of the basal forebrain in 167 non-demented carriers of the Colombian PSEN1 E280A mutation and 75 age- and sex-matched non-mutation carriers of the same kindred using a Bayesian analysis framework. We analyzed baseline MRI, amyloid PET, and FDG-PET scans of the Alzheimer's Prevention Initiative ADAD Colombia Trial. We found moderate evidence against an association of carrier status with basal forebrain volume (Bayes factor (BF10) = 0.182). We found moderate evidence against a difference of basal forebrain metabolism (BF10 = 0.167). There was only inconclusive evidence for an association between basal forebrain volume and delayed memory and attention (BF10 = 0.884 and 0.184, respectively), and between basal forebrain volume and global amyloid load (BF10 = 2.1). Our results distinguish PSEN1 E280A mutation carriers from sporadic AD cases in which cholinergic involvement of the basal forebrain is already detectable in the preclinical and prodromal stages. This indicates an important difference between ADAD and sporadic AD in terms of pathogenesis and potential treatment targets.


Sujet(s)
Maladie d'Alzheimer , Prosencéphale basal , Hétérozygote , Mutation , Tomographie par émission de positons , Préséniline-1 , Humains , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/métabolisme , Femelle , Mâle , Préséniline-1/génétique , Adulte d'âge moyen , Colombie , Prosencéphale basal/métabolisme , Prosencéphale basal/anatomopathologie , Prosencéphale basal/imagerie diagnostique , Imagerie par résonance magnétique , Adulte , Atrophie , Sujet âgé , Théorème de Bayes
17.
Int J Mol Sci ; 25(8)2024 Apr 11.
Article de Anglais | MEDLINE | ID: mdl-38673848

RÉSUMÉ

Alzheimer's disease is associated with protein aggregation, oxidative stress, and the role of acetylcholinesterase in the pathology of the disease. Previous investigations have demonstrated that geniposide and harpagoside protect the brain neurons, and cerium nanoparticles (CeO2 NPs) have potent redox and antioxidant properties. Thus, the effect of nanoparticles of Ce NPs and geniposide and harpagoside (GH/CeO2 NPs) on ameliorating AD pathogenesis was established on AlCl3-induced AD in mice and an aggregation proteins test in vitro. Findings of spectroscopy analysis have revealed that GH/CeO2 NPs are highly stable, nano-size, spherical in shape, amorphous nature, and a total encapsulation of GH in cerium. Treatments with CeO2 NPs, GH/CeO2 NPs, and donepezil used as positive control inhibit fibril formation and protein aggregation, protect structural modifications in the BSA-ribose system, have the ability to counteract Tau protein aggregation and amyloid-ß1-42 aggregation under fibrillation condition, and are able to inhibit AChE and BuChE. While the GH/CeO2 NPs, treatment in AD induced by AlCl3 inhibited amyloid-ß1-42, substantially enhanced the memory, the cognition coordination of movement in part AD pathogenesis may be alleviated through reducing amyloidogenic pathway and AChE and BuChE activities. The findings of this work provide important comprehension of the chemoprotective activities of iridoids combined with nanoparticles. This could be useful in the development of new therapeutic methods for the treatment of neurodegenerative diseases.


Sujet(s)
Acetylcholinesterase , Maladie d'Alzheimer , Cérium , Iridoïdes , Neuroprotecteurs , Cérium/composition chimique , Cérium/pharmacologie , Iridoïdes/pharmacologie , Iridoïdes/composition chimique , Animaux , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/composition chimique , Souris , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Acetylcholinesterase/métabolisme , Peptides bêta-amyloïdes/métabolisme , Mâle , Nanoparticules/composition chimique , Nanoparticules métalliques/composition chimique , Modèles animaux de maladie humaine
18.
Neurotox Res ; 42(2): 23, 2024 Apr 05.
Article de Anglais | MEDLINE | ID: mdl-38578482

RÉSUMÉ

Alzheimer's disease (AD) involves a neurodegenerative process that has not yet been prevented, reversed, or stopped. Continuing with the search for natural pharmacological treatments, flavonoids are a family of compounds with proven neuroprotective effects and multi-targeting behavior. The American genus Dalea L. (Fabaceae) is an important source of bioactive flavonoids. In this opportunity, we tested the neuroprotective potential of three prenylated flavanones isolated from Dalea species in a new in vitro pre-clinical AD model previously developed by us. Our approach consisted in exposing neural cells to conditioned media (3xTg-AD ACM) from neurotoxic astrocytes derived from hippocampi and cortices of old 3xTg-AD mice, mimicking a local neurodegenerative microenvironment. Flavanone 1 and 3 showed a neuroprotective effect against 3xTg-AD ACM, being 1 more active than 3. The structural requirements to afford neuroprotective activity in this model are a 5'-dimethylallyl and 4'-hydroxy at the B ring. In order to search the mechanistic performance of the most active flavanone, we focus on the flavonoid-mediated regulation of GSK-3ß-mediated tau phosphorylation previously reported. Flavanone 1 treatment decreased the rise of hyperphosphorylated tau protein neuronal levels induced after 3xTg-AD ACM exposure and inhibited the activity of GSK-3ß. Finally, direct exposure of these neurotoxic 3xTg-AD astrocytes to flavanone 1 resulted in toxicity to these cells and reduced the neurotoxicity of 3xTg-AD ACM as well. Our results allow us to present compound 1 as a natural prenylated flavanone that could be used as a precursor to development and design of future drug therapies for AD.


Sujet(s)
Maladie d'Alzheimer , Flavanones , Neuroprotecteurs , Souris , Animaux , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Glycogen synthase kinase 3 beta/métabolisme , Souris transgéniques , Protéines tau/métabolisme , Flavanones/pharmacologie , Flavanones/usage thérapeutique , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Modèles animaux de maladie humaine , Phosphorylation , Peptides bêta-amyloïdes/métabolisme
20.
Free Radic Biol Med ; 217: 141-156, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38552927

RÉSUMÉ

Current studies indicate that pathological modifications of tau are associated with mitochondrial dysfunction, synaptic failure, and cognitive decline in neurological disorders and aging. We previously showed that caspase-3 cleaved tau, a relevant tau form in Alzheimer's disease (AD), affects mitochondrial bioenergetics, dynamics and synaptic plasticity by the opening of mitochondrial permeability transition pore (mPTP). Also, genetic ablation of tau promotes mitochondrial function boost and increased cognitive capacities in aging mice. However, the mechanisms and relevance of these alterations for the cognitive and mitochondrial abnormalities during aging, which is the primary risk factor for AD, has not been explored. Therefore, in this study we used aging C57BL/6 mice (2-15 and 28-month-old) to evaluate hippocampus-dependent cognitive performance and mitochondrial function. Behavioral tests revealed that aged mice (15 and 28-month-old) showed a reduced cognitive performance compared to young mice (2 month). Concomitantly, isolated hippocampal mitochondria of aged mice showed a significant decrease in bioenergetic-related functions including increases in reactive oxygen species (ROS), mitochondrial depolarization, ATP decreases, and calcium handling defects. Importantly, full-length and caspase-3 cleaved tau were preferentially present in mitochondrial fractions of 15 and 28-month-old mice. Also, aged mice (15 and 28-month-old) showed an increase in cyclophilin D (CypD), the principal regulator of mPTP opening, and a decrease in Opa-1 mitochondrial localization, indicating a possible defect in mitochondrial dynamics. Importantly, we corroborated these findings in immortalized cortical neurons expressing mitochondrial targeted full-length (GFP-T4-OMP25) and caspase-3 cleaved tau (GFP-T4C3-OMP25) which resulted in increased ROS levels and mitochondrial fragmentation, along with a decrease in Opa-1 protein expression. These results suggest that tau associates with mitochondria and this binding increases during aging. This connection may contribute to defects in mitochondrial bioenergetics and dynamics which later may conduce to cognitive decline present during aging.


Sujet(s)
Maladie d'Alzheimer , Dysfonctionnement cognitif , Souris , Animaux , Espèces réactives de l'oxygène/métabolisme , Caspase-3/métabolisme , Souris de lignée C57BL , Maladie d'Alzheimer/métabolisme , Dysfonctionnement cognitif/anatomopathologie , Vieillissement/génétique , Mitochondries/métabolisme , Hippocampe/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE