Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.768
Filtrar
1.
J Neuroinflammation ; 21(1): 215, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39218898

RESUMEN

BACKGROUND: Alzheimer's disease (AD) features progressive neurodegeneration and microglial activation that results in dementia and cognitive decline. The release of soluble amyloid (Aß) oligomers into the extracellular space is an early feature of AD pathology. This can promote excitotoxicity and microglial activation. Microglia can adopt several activation states with various functional outcomes. Protective microglial activation states have been identified in response to Aß plaque pathology in vivo. However, the role of microglia and immune mediators in neurotoxicity induced by soluble Aß oligomers is unclear. Further, there remains a need to identify druggable molecular targets that promote protective microglial states to slow or prevent the progression of AD. METHODS: Hippocampal entorhinal brain slice culture (HEBSC) was employed to study mechanisms of Aß1-42 oligomer-induced neurotoxicity as well as the role of microglia. The roles of glutamate hyperexcitation and immune signaling in Aß-induced neurotoxicity were assessed using MK801 and neutralizing antibodies to the TNF-related apoptosis-inducing ligand (TRAIL) respectively. Microglial activation state was manipulated using Gi-hM4di designer receptor exclusively activated by designer drugs (DREADDs), microglial depletion with the colony-stimulating factor 1 receptor (CSF1R) antagonist PLX3397, and microglial repopulation (PLX3397 withdrawal). Proteomic changes were assessed by LC-MS/MS in microglia isolated from control, repopulated, or Aß-treated HEBSCs. RESULTS: Neurotoxicity induced by soluble Aß1-42 oligomers involves glutamatergic hyperexcitation caused by the proinflammatory mediator and death receptor ligand TRAIL. Microglia were found to have the ability to both promote and restrain Aß-induced toxicity. Induction of microglial Gi-signaling with hM4di to prevent pro-inflammatory activation blunted Aß neurotoxicity, while microglial depletion with CSF1R antagonism worsened neurotoxicity caused by Aß as well as TRAIL. HEBSCs with repopulated microglia, however, showed a near complete resistance to Aß-induced neurotoxicity. Comparison of microglial proteomes revealed that repopulated microglia have a baseline anti-inflammatory and trophic phenotype with a predicted pathway activation that is nearly opposite that of Aß-exposed microglia. mTORC2 and IRF7 were identified as potential targets for intervention. CONCLUSION: Microglia are key mediators of both protection and neurodegeneration in response to Aß. Polarizing microglia toward a protective state could be used as a preventative strategy against Aß-induced neurotoxicity.


Asunto(s)
Péptidos beta-Amiloides , Microglía , Fragmentos de Péptidos , Ligando Inductor de Apoptosis Relacionado con TNF , Microglía/metabolismo , Microglía/efectos de los fármacos , Péptidos beta-Amiloides/toxicidad , Péptidos beta-Amiloides/metabolismo , Animales , Fragmentos de Péptidos/toxicidad , Fragmentos de Péptidos/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/toxicidad , Ratones , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Ratones Endogámicos C57BL , Corteza Entorrinal/metabolismo , Corteza Entorrinal/efectos de los fármacos , Corteza Entorrinal/patología , Técnicas de Cultivo de Órganos
2.
CNS Neurosci Ther ; 30(8): e14878, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39097923

RESUMEN

BACKGROUND: This work elucidated the effect of honokiol (HKL) on hippocampal neuronal mitochondrial function in Alzheimer's disease (AD). METHODS: APP/PS1 mice were used as AD mice models and exposed to HKL and 3-TYP. Morris water maze experiment was performed to appraise cognitive performance of mice. Hippocampal Aß+ plaque deposition and neuronal survival was evaluated by immunohistochemistry and Nissl staining. Hippocampal neurons were dissociated from C57BL/6 mouse embryos. Hippocampal neuronal AD model was constructed by Aß oligomers induction and treated with HKL, CsA and 3-TYP. Neuronal viability and apoptosis were detected by cell counting kit-8 assay and TUNEL staining. mRFP-eGFP-LC3 assay, MitoSOX Red, dichlorodihydrofluorescein diacetate, and JC-1 staining were performed to monitor neuronal autophagosomes, mitochondrial reactive oxygen species (ROS), neuronal ROS, and mitochondrial membrane potential. Autophagy-related proteins were detected by Western blot. RESULTS: In AD mice, HKL improved cognitive function, relieved hippocampal Aß1-42 plaque deposition, promoted hippocampal neuron survival, and activated hippocampal SIRT3 expression and mitochondrial autophagy. These effects of HKL on AD mice were abolished by 3-TYP treatment. In hippocampal neuronal AD model, HKL increased neuronal activity, attenuated neuronal apoptosis and Aß aggregation, activated SIRT3 and mitochondrial autophagy, reduced mitochondrial and neuronal ROS, and elevated mitochondrial membrane potential. CsA treatment and 3-TYP treatment abrogated the protection of HKL on hippocampal neuronal AD model. The promotion of mitochondrial autophagy by HKL in hippocampal neuronal AD model was counteracted by 3-TYP. CONCLUSIONS: HKL activates SIRT3-mediated mitochondrial autophagy to mitigate hippocampal neuronal damage in AD. HKL may be effective in treating AD.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Autofagia , Compuestos de Bifenilo , Hipocampo , Lignanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias , Neuronas , Sirtuina 3 , Animales , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/patología , Hipocampo/metabolismo , Sirtuina 3/metabolismo , Ratones , Neuronas/efectos de los fármacos , Neuronas/patología , Neuronas/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Compuestos de Bifenilo/farmacología , Autofagia/efectos de los fármacos , Lignanos/farmacología , Péptidos beta-Amiloides/toxicidad , Fragmentos de Péptidos/toxicidad , Masculino , Fármacos Neuroprotectores/farmacología , Modelos Animales de Enfermedad , Especies Reactivas de Oxígeno/metabolismo , Compuestos Alílicos , Fenoles
3.
Physiol Behav ; 285: 114652, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39096985

RESUMEN

Patients with Alzheimer's disease (AD) present with a variety of symptoms, including core symptoms as well as behavioral and psychological symptoms. Somatosensory neural systems are generally believed to be relatively unaffected by AD until late in the course of the disease; however, somatosensory perception in patients with AD is not yet well understood. One factor that may complicate the assessment of somatosensory perception in humans centers on individual variations in pathological and psychological backgrounds. It is therefore necessary to evaluate somatosensory perception using animal models with uniform status. In the current study, we focused on the hippocampus, the primary site of AD. We first constructed a rat model of AD model using bilateral hippocampal injections of amyloid-ß peptide 1-40 and ibotenic acid; sham rats received saline injections. The Morris water maze test was used to evaluate memory impairment, and the formalin test (1 % or 4 % formalin) and upper lip von Frey test were performed to compare pain perception between AD model and sham rats. Finally, histological and immunohistochemical methods were used to evaluate tissue damage and neuronal activity, respectively, in the hippocampus. AD model rats showed bilateral hippocampal damage and had memory impairment in the Morris water maze test. Furthermore, AD model rats exhibited significantly less pain-related behavior in phase 2 (the last 50 min of the 60-minute observation) of the 4 % formalin test compared with the sham rats. However, no significant changes were observed in the von Frey test. Immunohistochemical observations of the trigeminal spinal subnucleus caudalis after 4 % formalin injection revealed significantly fewer c-Fos-immunoreactive cells in AD model rats than in sham rats, reflecting reduced neuronal activity. These results indicate that AD model rats with hippocampal damage have reduced responsiveness to persistent inflammatory chemical stimuli to the orofacial region.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Modelos Animales de Enfermedad , Hipocampo , Ácido Iboténico , Percepción del Dolor , Fragmentos de Péptidos , Ratas Sprague-Dawley , Animales , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/metabolismo , Ácido Iboténico/toxicidad , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/patología , Masculino , Percepción del Dolor/efectos de los fármacos , Percepción del Dolor/fisiología , Fragmentos de Péptidos/toxicidad , Ratas , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Dimensión del Dolor , Trastornos de la Memoria/etiología
4.
Neurochem Int ; 179: 105839, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39173832

RESUMEN

The intricate nature of Alzheimer's disease (AD) has presented significant hurdles in the development of effective interventions. Sulforaphane (SFN) is of interest due to its antioxidative, anti-inflammatory, and neuroprotective properties, which could address various aspects of AD pathology. This study explores the potential of SFN in a rat model of AD induced by Aß (1-42) peptides. AD symptoms were triggered in rats by injecting Aß (1-42) peptides directly into their cerebral ventricles. SFN (10 mg/kg and 20 mg/kg), Trigonelline (10 mg/kg), and Pioglitazone (10 mg/kg) were administered in Aß (1-42) treated animals. Behavioral assessments were performed using the Novel Object Recognition tests. Various biochemical parameters, such as soluble Aß (1-42), IRS-S312, GSK-3ß, TNF-α, acetylcholinesterase, nitrite levels, lipid peroxidation, and reduced glutathione activity, were quantified using ELISA kits and spectrophotometric assays. Histopathological analyses included Hematoxylin and Eosin, Crystal Violet, Congo red, and IRS-1 Immunohistochemistry staining. Quantification was performed to assess neuronal loss and Aß plaque burden. The novelty of this study lies in its comprehensive evaluation of SFN's impact on multiple AD-related pathways at dual doses. The Novel Object Recognition test revealed that SFN, especially at higher doses, improved memory deficits induced by Aß (1-42). Biochemically, SFN reduced hippocampal Aß levels, IRS-S312, GSK-3ß, TNF-α, and acetylcholinesterase activity, while increasing glutathione levels, all in a dose-dependent manner. Histopathological analyses further confirmed SFN's protective role against Aß-induced neuronal damage, amyloidosis, and changes in insulin signaling. These results highlight SFN's potential as a multifaceted therapeutic agent for AD, offering a promising avenue for treatment due to its antioxidative, anti-inflammatory, and neuroprotective properties. The inclusion of combination treatments with Trigonelline and Pioglitazone alongside SFN offers insights into potential synergistic effects, which could pave the way for developing combination therapies for AD.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Modelos Animales de Enfermedad , Isotiocianatos , Fármacos Neuroprotectores , Fragmentos de Péptidos , Sulfóxidos , Animales , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Isotiocianatos/farmacología , Isotiocianatos/uso terapéutico , Fragmentos de Péptidos/toxicidad , Masculino , Ratas , Sulfóxidos/farmacología , Ratas Wistar
5.
Int J Mol Sci ; 25(16)2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39201616

RESUMEN

Amyloid ß peptide (Aß) aggregation and deposition are considered the main causes of Alzheimer's disease. In a previous study, we demonstrated that anionic Zn-phthalocyanine (ZnPc) can interact with the Aß peptide and inhibit the fibril-formation process. However, due to the inability of anionic ZnPc to cross the intact blood-brain barrier, we decided to explore the interaction of cationic methylated Zn-phthalocyanine (cZnPc) with the peptide. Using a ThT fluorescence assay, we observed that cZnPc dose-dependently and time-dependently inhibited Aß1-42 fibril levels under in vitro fibril-formation conditions. Electron microscopy revealed that it caused Aß1-42 peptides to form small aggregates. Western blotting and dot immunoblot oligomer experiments demonstrated that cZnPc increased rather than decreased the levels of oligomers from the very early stages of incubation. A binding assay confirmed that cZnPc could bind with the peptide. Docking simulations indicated that the oligomer species of Aß1-42 had a higher ability to interact with cZnPc. ANS fluorescence assay results indicated that cZnPc did not affect the hydrophobicity of the peptide. However, cZnPc significantly increased intrinsic tyrosine fluorescence of the peptide after 8 h of incubation in fibril-formation conditions. Importantly, cell culture experiments demonstrated that cZnPc did not exhibit any toxicity up to a concentration of 10 µM. Instead, it protected a neuronal cell line from Aß1-42-induced toxicity. Thus, our results suggest that cZnPc can affect the aggregation process of Aß1-42, rendering it non-toxic, which could be crucial for the therapy of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Indoles , Isoindoles , Compuestos Organometálicos , Fragmentos de Péptidos , Compuestos de Zinc , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Indoles/química , Indoles/farmacología , Humanos , Compuestos de Zinc/química , Compuestos de Zinc/farmacología , Compuestos Organometálicos/farmacología , Compuestos Organometálicos/química , Fragmentos de Péptidos/química , Fragmentos de Péptidos/toxicidad , Fragmentos de Péptidos/farmacología , Agregado de Proteínas/efectos de los fármacos , Animales , Simulación del Acoplamiento Molecular , Neuronas/efectos de los fármacos , Neuronas/metabolismo
6.
J Alzheimers Dis ; 101(1): 49-60, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39093068

RESUMEN

Background: Recent advances linking gut dysbiosis with neurocognitive disorders such as Alzheimer's disease (AD) suggest that the microbiota-gut-brain axis could be targeted for AD prevention, management, or treatment. Objective: We sought to identify probiotics that can delay Aß-induced paralysis. Methods: Using C. elegans expressing human amyloid-ß (Aß)1-42 in body wall muscles (GMC101), we assessed the effects of several probiotic strains on paralysis. Results: We found that Lacticaseibacillus rhamnosus HA-114 and Bacillus subtilis R0179, but not their supernatants or heat-treated forms, delayed paralysis and prolonged lifespan without affecting the levels of amyloid-ß aggregates. To uncover the mechanism involved, we explored the role of two known pathways involved in neurogenerative diseases, namely mitophagy, via deletion of the mitophagy factor PINK-1, and fatty acid desaturation, via deletion of the Δ9 desaturase FAT-5. Pink-1 deletion in GMC101 worms did not modify the life-prolonging and anti-paralysis effects of HA-114 but reduced the protective effect of R0179 against paralysis without affecting its life-prolonging effect. Upon fat5 deletion in GMC101 worms, the monounsaturated C14:1 and C16:1 FAs conserved their beneficial effect while the saturated C14:0 and C16:0 FAs did not. The beneficial effects of R0179 on both lifespan and paralysis remained unaffected by fat-5 deletion, while the beneficial effect of HA-114 on paralysis and lifespan was significantly reduced. Conclusions: Collectively with clinical and preclinical evidence in other models, our results suggest that HA-114 or R0179 could be studied as potential therapeutical adjuncts in neurodegenerative diseases such as AD.


Asunto(s)
Péptidos beta-Amiloides , Bacillus subtilis , Caenorhabditis elegans , Lacticaseibacillus rhamnosus , Longevidad , Probióticos , Animales , Longevidad/efectos de los fármacos , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Parálisis , Fragmentos de Péptidos/toxicidad , Fragmentos de Péptidos/metabolismo , Animales Modificados Genéticamente , Humanos , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo
7.
Neurotox Res ; 42(4): 37, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-39102123

RESUMEN

Amyloid-peptide (Aß) monomeric forms (ABM) occurring in presymptomatic Alzheimer's disease (AD) brain are thought to be devoid of neurotoxicity while the transition/aggregation of ABM into oligomers is determinant for Aß-induced toxicity since Aß is predominantly monomeric up to 3 µM and aggregates over this concentration. However, recent imaging and/or histopathological investigations revealed alterations of myelin in prodromal AD brain in absence of aggregated Aß oligomers, suggesting that ABM may induce toxicity in myelin-producing cells in early AD-stages. To check this hypothesis, here we studied ABM effects on the viability of the Human oligodendrocyte cell line (HOG), a reliable oligodendrocyte model producing myelin proteins. Furthermore, to mimic closely interactions between oligodendrocytes and other glial cells regulating myelination, we investigated also ABM effects on mouse brain primary mixed-glial cell cultures. Various methods were combined to show that ABM concentrations (600 nM-1 µM), extremely lower than 3 µM, significantly decreased HOG cell and mouse brain primary mixed-glial cell survival. Interestingly, flow-cytometry studies using specific cell-type markers demonstrated that oligodendrocytes represent the most vulnerable glial cell population affected by ABM toxicity. Our work also shows that the neurosteroid 3α-O-allyl-allopregnanolone BR351 (250 and 500 nM) efficiently prevented ABM-induced HOG and brain primary glial cell toxicity. Bicuculline (50-100 nM), the GABA-A-receptor antagonist, was unable to block/reduce BR351 effect against ABM-induced HOG and primary glial cell toxicity, suggesting that BR351-evoked neuroprotection of these cells may not depend on GABA-A-receptor allosterically modulated by neurosteroids. Altogether, our results suggest that further exploration of BR351 therapeutic potential may offer interesting perspectives to develop effective neuroprotective strategies.


Asunto(s)
Péptidos beta-Amiloides , Fármacos Neuroprotectores , Oligodendroglía , Pregnanolona , Animales , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Humanos , Péptidos beta-Amiloides/toxicidad , Fármacos Neuroprotectores/farmacología , Pregnanolona/farmacología , Ratones , Línea Celular , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Ratones Endogámicos C57BL , Fragmentos de Péptidos/toxicidad , Células Cultivadas , Relación Dosis-Respuesta a Droga
8.
Brain Res Bull ; 215: 111030, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38996935

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative brain disorder that progressively impairs long-term and working memory. The function and mechanism of PA(Patchouli alcohol) in improving AD in the external treatment of encephalopathy remain unclear. This study aimed to investigate the therapeutic effect of PA on AD using an Aß1-42 induced AD mouse model with LPS(Lipopolysaccharide) stimulation of BV2 microglial cells. Additionally, we aimed to explore the potential mechanism of PA in enhancing autophagy and reducing neuroinflammation through the AMPK (AMP-activated protein kinase)/mTOR (Mammaliam target of rapamycin) signaling pathway. The Morris water maze was used to assess cognitive function, and cortical and hippocampal tissues were collected for further analysis of the corresponding signaling pathways and inflammatory changes through biological experiments. Our research findings demonstrate that PA has a significant positive impact on cognitive and memory impairments in mice that have been induced with Aß1-42-induced AD. Additionally, PA was also found to revert the activation of microglia induced by LPS. These effects may be attributed to the reduction of neuroinflammation and enhancement of the AMPK/mTOR autophagy pathway. Therefore, PA may serve as an effective therapeutic option to prevent or delay the progression of AD-associated memory dysfunction.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Enfermedad de Alzheimer , Péptidos beta-Amiloides , Disfunción Cognitiva , Microglía , Fragmentos de Péptidos , Transducción de Señal , Serina-Treonina Quinasas TOR , Animales , Péptidos beta-Amiloides/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Ratones , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Proteínas Quinasas Activadas por AMP/metabolismo , Transducción de Señal/efectos de los fármacos , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Modelos Animales de Enfermedad
9.
AAPS PharmSciTech ; 25(6): 149, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38954224

RESUMEN

Silibinin (SIL) Encapsulated Nanoliquid Crystalline (SIL-NLCs) particles were prepared to study neuroprotective effect against amyloid beta (Aß1-42) neurotoxicity in Balb/c mice model. Theses NLCs were prepared through hot emulsification and probe sonication technique. The pharmacodynamics was investigatigated on Aß1-42 intracerebroventricular (ICV) injected Balb/c mice. The particle size, zeta potential and drug loading were optimized to be 153 ± 2.5 nm, -21 mV, and 8.2%, respectively. Small angle X-ray (SAXS) and electron microscopy revealed to crystalline shape of SIL-NLCs. Thioflavin T (ThT) fluroscence and circular dichroism (CD) technique were employed to understand monomer inhibition effect of SIL-NLCs on Aß1-4. In neurobehavioral studies, SIL-NLCs exhibited enhanced mitigation of memory impairment induced on by Aß1-42 in T-maze and new object recognition test (NORT). Whereas biochemical and histopathological estimation of brain samples showed reduction in level of Aß1-42 aggregate, acetylcholine esterase (ACHE) and reactive oxygen species (ROS). SIL-NLCs treated animal group showed higher protection against Aß1-42 toxicity compared to free SIL and Donopezil (DPZ). Therefore SIL-NLCs promises great prospect in neurodegenerative diseases such as Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides , Ratones Endogámicos BALB C , Fármacos Neuroprotectores , Fragmentos de Péptidos , Silibina , Animales , Péptidos beta-Amiloides/toxicidad , Péptidos beta-Amiloides/metabolismo , Ratones , Silibina/farmacología , Silibina/administración & dosificación , Fragmentos de Péptidos/toxicidad , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/administración & dosificación , Masculino , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Tamaño de la Partícula , Nanopartículas/química , Especies Reactivas de Oxígeno/metabolismo , Modelos Animales de Enfermedad , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Acetilcolinesterasa/metabolismo
10.
Brain Res ; 1842: 149102, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-38969084

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disorder. Accumulation of ß-amyloid (Aß) in the brain has been recognized as a key factor in the onset and progression of Alzheimer's disease (AD).The accumulation of Aß in the brain catalyzes the production of reactive oxygen species (ROS), which in turn triggers oxidative damage to cellular components such as DNA, lipids, and proteins. In the present study, we investigated the protective effect of Ganoderic acid A (GA.A) against Aß42-induced apoptosis in PC12 cells. Changes in mitochondrial membrane potential indicated that GA.A treats mitochondrial dysfunction by decreasing Aß42 deposition and inhibiting neural protofiber tangle formation. Changes in intracellular Ca2+ and caspase-3 indicated that GA.A reduced mitochondrial damage by Aß42 in PC12 cells, thereby decreasing ROS accumulation and reducing Aß protofiber-induced cytotoxicity. These features suggest that GA.A has great potential as an effective neuroprotective drug in the treatment of Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides , Apoptosis , Lanosterol , Potencial de la Membrana Mitocondrial , Mitocondrias , Fármacos Neuroprotectores , Fragmentos de Péptidos , Especies Reactivas de Oxígeno , Animales , Péptidos beta-Amiloides/toxicidad , Péptidos beta-Amiloides/metabolismo , Células PC12 , Ratas , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Fragmentos de Péptidos/toxicidad , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Apoptosis/efectos de los fármacos , Lanosterol/farmacología , Lanosterol/análogos & derivados , Calcio/metabolismo , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Ácidos Heptanoicos
11.
ACS Chem Neurosci ; 15(15): 2916-2924, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39036818

RESUMEN

Several studies have highlighted the presence of nitration damage following neuroinflammation in Alzheimer's disease (AD). Accordingly, post-transcriptional modifications of ß-amyloid (Aß), including peptide nitration, have been explored as a marker of the disease. However, the implications of Aß nitration in terms of aggregation propensity and neurotoxicity are still debated. Here, we show new data obtained using a photoactivatable peroxynitrite generator (BPT-NO) to overcome the limitations associated with chemical nitration methods. We found that the photoactivation of BPT-NO with the highly biocompatible red light selectively induces the nitration of tyrosine 10 of freshly solubilized full-length Aß1-42. Photonitrated Aß1-42 was, therefore, investigated for aggregation states and functions. It resulted that photonitrated Aß1-42 did not aggregate into small oligomers but rather self-assembled into large amorphous aggregates. When tested on neuronal-like SH-SY5Y cells and microglial C57BL/6 BV2 cells, photonitrated Aß1-42 showed to be free of neurotoxicity and able to induce phagocytic microglia cells. We propose that light-controlled nitration of the multiple forms in which Aß occurs (i.e., monomers, oligomers, fibrils) could be a tool to assess in real-time the impact of tyrosine nitration on the amyloidogenic and toxic properties of Aß1-42.


Asunto(s)
Péptidos beta-Amiloides , Luz , Fragmentos de Péptidos , Tirosina , Péptidos beta-Amiloides/metabolismo , Tirosina/metabolismo , Tirosina/análogos & derivados , Tirosina/química , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Humanos , Animales , Microglía/metabolismo , Microglía/efectos de los fármacos , Ácido Peroxinitroso/metabolismo , Ratones , Agregado de Proteínas/fisiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Neuronas/metabolismo , Neuronas/efectos de los fármacos
12.
Biochem Pharmacol ; 226: 116409, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38969300

RESUMEN

Epilepsy is a central nervous system (CNS) disorder causing repeated seizures due to a transient excessive or synchronous alteration in the electrical activity of the brain. Several neurological disorders have been associated to gluten-related diseases (GRD), including epilepsy. However, the molecular mechanisms that associate GRD and epileptogenesis are still unknown. Our previous data have shown that the gliadin peptide 31-43 (p31-43) enhanced number and duration of seizures induced by kainate in mice and exacerbated CA3-kainate-induced neurotoxicity in organotypic hippocampal slices. Here, we investigated whether another important gliadin peptide p57-68 may exerts effects similar to p31-43 on kainate-induced neurotoxicity. We find that both peptides exacerbate kainate-induced damage in the CA3 region once simultaneously challenged. However, after pre-incubation, p31-43 additionally exacerbates neurotoxicity in the CA1 region, while p57-68 does not. These data suggested differential intracellular mechanisms activated by the peptides. Indeed, analysing intracellular signalling pathways we discover that p31-43 induces significant intracellular changes, including increased phosphorylation of Akt, Erk1/2, and p65, decreased p38 phosphorylation, and deacetylation of nuclear histone-3. Based on these observations, we demonstrate that p31-43 likely activates specific intracellular signaling pathways involved in neuronal excitability, inflammation, and epigenetic regulation, which may contribute to its exacerbation of kainate-induced neurotoxicity. In contrast, p57-68 appears to exert its effects through different mechanisms. Further research is necessary to elucidate the precise mechanisms by which these peptides influence neurotoxicity and understand their implications for neurological disorders.


Asunto(s)
Epilepsia , Gliadina , Animales , Epilepsia/metabolismo , Epilepsia/inducido químicamente , Gliadina/toxicidad , Gliadina/metabolismo , Fragmentos de Péptidos/toxicidad , Fragmentos de Péptidos/metabolismo , Ácido Kaínico/toxicidad , Ratones , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos
13.
Toxicol In Vitro ; 99: 105880, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38901785

RESUMEN

Alzheimer's disease (AD), the most prevalent form of dementia worldwide, is a significant health concern, according to the World Health Organization (WHO). The neuropathological diagnostic criteria for AD are based on the deposition of amyloid-ß peptide (Aß) and the formation of intracellular tau protein tangles. These proteins are associated with several overlapping neurodegenerative mechanisms, including oxidative stress, mitochondrial dysfunction, lipid peroxidation, reduced neuronal viability, and cell death. In this context, our study focuses on the potential therapeutic use of cannabidiol (CBD), a non-psychotropic cannabinoid with antioxidant and anti-inflammatory effects. We aim to evaluate CBD's neuroprotective role, particularly in protecting hippocampal neurons from Aß25-35-induced toxicity. Our findings indicate that CBD significantly improves cell viability and decreases levels of lipid peroxidation and oxidative stress. The results demonstrate that CBD possesses a robust potential to rescue cells from induced neurotoxicity through its antioxidant properties. Additionally, the neuroprotective effect of CBD may be associated with the modulation of the endocannabinoid system. These findings suggest that CBD could be a promising compound for adjuvant treatments in neurodegenerative processes triggered by amyloid-ß peptide.


Asunto(s)
Péptidos beta-Amiloides , Cannabidiol , Supervivencia Celular , Hipocampo , Peroxidación de Lípido , Neuronas , Fármacos Neuroprotectores , Estrés Oxidativo , Fragmentos de Péptidos , Péptidos beta-Amiloides/toxicidad , Cannabidiol/farmacología , Animales , Fármacos Neuroprotectores/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fragmentos de Péptidos/toxicidad , Hipocampo/efectos de los fármacos , Hipocampo/citología , Hipocampo/metabolismo , Ratones , Supervivencia Celular/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Células Cultivadas , Antioxidantes/farmacología , Especies Reactivas de Oxígeno/metabolismo
14.
CNS Neurosci Ther ; 30(6): e14814, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38887858

RESUMEN

AIMS: Alzheimer's disease (AD) is a devastating dementia characterized by extracellular amyloid-ß (Aß) protein aggregates and intracellular tau protein deposition. Clinically available drugs mainly target acetylcholinesterase (AChE) and indirectly sustain cholinergic neuronal tonus. Butyrylcholinesterase (BChE) also controls acetylcholine (ACh) turnover and is involved in the formation of Aß aggregates and senile plaques. UW-MD-95 is a novel carbamate-based compound acting as a potent pseudo-irreversible BChE inhibitor, with high selectivity versus AChE, and showing promising protective potentials in AD. METHODS: We characterized the neuroprotective activity of UW-MD-95 in mice treated intracerebroventricularly with oligomerized Aß25-35 peptide using behavioral, biochemical, and immunohistochemical approaches. RESULTS: When injected acutely 30 min before the behavioral tests (spontaneous alternation in the Y-maze, object recognition, or passive avoidance), UW-MD-95 (0.3-3 mg/kg) showed anti-amnesic effects in Aß25-35-treated mice. When injected once a day over 7 days, it prevented Aß25-35-induced memory deficits. This effect was lost in BChE knockout mice. Moreover, the compound prevented Aß25-35-induced oxidative stress (assessed by lipid peroxidation or cytochrome c release), neuroinflammation (IL-6 and TNFα levels or GFAP and IBA1 immunoreactivity) in the hippocampus and cortex, and apoptosis (Bax level). Moreover, UW-MD-95 significantly reduced the increase in soluble Aß1-42 level in the hippocampus induced by Aß25-35. CONCLUSION: UW-MD-95 appeared as a potent neuroprotective compound in the Aß25-35 model of AD, with potentially an impact on Aß1-42 accumulation that could suggest a novel mechanism of neuroprotection.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Butirilcolinesterasa , Inhibidores de la Colinesterasa , Modelos Animales de Enfermedad , Fármacos Neuroprotectores , Fragmentos de Péptidos , Animales , Fármacos Neuroprotectores/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Ratones , Fragmentos de Péptidos/toxicidad , Masculino , Inhibidores de la Colinesterasa/farmacología , Butirilcolinesterasa/metabolismo , Ratones Endogámicos C57BL , Aprendizaje por Laberinto/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Estrés Oxidativo/efectos de los fármacos
15.
ACS Chem Neurosci ; 15(12): 2372-2385, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38822790

RESUMEN

Spontaneous aggregation of amyloid beta (Aß) leads to the formation of neurotoxic senile plaque considered as the most crucial event in Alzheimer's disease (AD) progression. Inhibition or disruption of this deadly aggregate formation is one of the most efficient strategies for the development of potential therapeutics, and extensive research is in progress by various research groups. In this direction, the development of a peptide analogous to that of the native Aß peptide is an attractive strategy. Based on this rationale, ß-sheet breakers were developed from the Aß central hydrophobic core. These peptide derivatives will bind to the full length of the parent Aß and interfere in self-recognition, thereby preventing the folding of the Aß peptide into cross ß-sheet neurotoxic aggregates. However, this approach is effective in the inhibition of fibrillar aggregation, but this strategy is ineffective in the Aß neurotoxic oligomer formation. Therefore, an alternative and efficient approach is to use the Aß peptide analogous to the C-terminal region, which arbitrates fibrillation and oligomerization. Herein, we developed the Aß C-terminal fragment (ACT-1 to ACT-7) for inhibition of oligomerization as well as fibrillar aggregation. Screening of these seven peptides resulted in an efficient anti-Aß peptide aggregative agent (ACT-7), which was evaluated by the ThT assay peptide. The ThT assay reveals complete inhibition and showed significant neuroprotection of PC-12-derived neurons from Aß-induced toxicity and reduced cell apoptosis. Further, analysis using CD and FTIR spectroscopy reveals that the ACT-7 peptide efficiently inhibits the formation of the ß-sheet secondary structure content. HR-TEM microscopic analysis confirmed the inhibition of formation. Therefore, the inhibition of ß-sheet Aß fibrillary aggregation by the protease-stable ACT-7 peptide may provide a beneficial effect on AD treatment to control the Aß aggregates. Finally, we anticipate that our newly designed ACT peptides may also assist as a template molecular scaffold for designing potential anti-AD therapeutics.


Asunto(s)
Péptidos beta-Amiloides , Neuronas , Fármacos Neuroprotectores , Fragmentos de Péptidos , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fragmentos de Péptidos/toxicidad , Fragmentos de Péptidos/farmacología , Animales , Fármacos Neuroprotectores/farmacología , Interacciones Hidrofóbicas e Hidrofílicas , Humanos , Ratas
16.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1869(6): 159510, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38759921

RESUMEN

Abrupt aggregation of amyloid ß1-42 (Aß1-42) peptide in the frontal lobe is the expected underlying cause of Alzheimer's disease (AD). ß-Sheet-rich oligomers and fibrils formed by Aß1-42 exert high cell toxicity. A growing body of evidence indicates that lipids can uniquely alter the secondary structure and toxicity of Aß1-42 aggregates. At the same time, underlying molecular mechanisms that determine this difference in toxicity of amyloid aggregates remain unclear. Using a set of molecular and biophysical assays to determine the molecular mechanism by which Aß1-42 aggregates formed in the presence of cholesterol, cardiolipin, and phosphatidylcholine exert cell toxicity. Our findings demonstrate that rat neuronal cells exposed to Aß1-42 fibrils formed in the presence of lipids with different chemical structure exert drastically different magnitude and dynamic of unfolded protein response (UPR) in the endoplasmic reticulum (ER) and mitochondria (MT). We found that the opposite dynamics of UPR in MT and ER in the cells exposed to Aß1-42: cardiolipin fibrils and Aß1-42 aggregates formed in a lipid-free environment. We also found that Aß1-42: phosphatidylcholine fibrils upregulated ER UPR simultaneously downregulating the UPR response of MT, whereas Aß1-42: cholesterol fibrils suppressed the UPR response of ER and upregulated UPR response of MT. We also observed progressively increasing ROS production that damages mitochondrial membranes and other cell organelles, ultimately leading to cell death.


Asunto(s)
Péptidos beta-Amiloides , Mitocondrias , Fragmentos de Péptidos , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Animales , Ratas , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Respuesta de Proteína Desplegada/efectos de los fármacos , Cardiolipinas/metabolismo , Colesterol/metabolismo , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Amiloide/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Fosfatidilcolinas/metabolismo , Fosfatidilcolinas/química , Humanos , Especies Reactivas de Oxígeno/metabolismo
17.
J Neuroinflammation ; 21(1): 144, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38822334

RESUMEN

Cumulative evidence has established that Interferon (IFN)-γ has both pathogenic and protective roles in Multiple Sclerosis and the animal model, Experimental Autoimmune Encephalomyelitis (EAE). However, the underlying mechanisms to the beneficial effects of IFN-γ are not well understood. In this study, we found that IFN-γ exerts therapeutic effects on chronic, relapsing-remitting, and chronic progressive EAE models. The frequency of regulatory T (Treg) cells in spinal cords from chronic EAE mice treated with IFN-γ was significantly increased with no effect on Th1 and Th17 cells. Consistently, depletion of FOXP3-expressing cells blocked the protective effects of IFN-γ, indicating that the therapeutic effect of IFN-γ depends on the presence of Treg cells. However, IFN-γ did not trigger direct in vitro differentiation of Treg cells. In vivo administration of blocking antibodies against either interleukin (IL)-10, transforming growth factor (TGF)-ß or program death (PD)-1, revealed that the protective effects of IFN-γ in EAE were also dependent on TGF-ß and PD-1, but not on IL-10, suggesting that IFN-γ might have an indirect role on Treg cells acting through antigen-presenting cells. Indeed, IFN-γ treatment increased the frequency of a subset of splenic CD11b+ myeloid cells expressing TGF-ß-Latency Associated Peptide (LAP) and program death ligand 1 (PD-L1) in a signal transducer and activator of transcription (STAT)-1-dependent manner. Furthermore, splenic CD11b+ cells from EAE mice preconditioned in vitro with IFN-γ and myelin oligodendrocyte glycoprotein (MOG) peptide exhibited a tolerogenic phenotype with the capability to induce conversion of naïve CD4+ T cells mediated by secretion of TGF-ß. Remarkably, adoptive transfer of splenic CD11b+ cells from IFN-γ-treated EAE mice into untreated recipient mice ameliorated clinical symptoms of EAE and limited central nervous system infiltration of mononuclear cells and effector helper T cells. These results reveal a novel cellular and molecular mechanism whereby IFN-γ promotes beneficial effects in EAE by endowing splenic CD11b+ myeloid cells with tolerogenic and therapeutic activities.


Asunto(s)
Antígeno CD11b , Encefalomielitis Autoinmune Experimental , Interferón gamma , Ratones Endogámicos C57BL , Células Mieloides , Bazo , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Ratones , Interferón gamma/metabolismo , Células Mieloides/efectos de los fármacos , Células Mieloides/inmunología , Células Mieloides/metabolismo , Bazo/inmunología , Antígeno CD11b/metabolismo , Femenino , Glicoproteína Mielina-Oligodendrócito/toxicidad , Glicoproteína Mielina-Oligodendrócito/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Fragmentos de Péptidos/toxicidad , Fragmentos de Péptidos/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/inmunología , Factores de Transcripción Forkhead/metabolismo , Modelos Animales de Enfermedad
18.
Molecules ; 29(9)2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38731472

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by the accumulation of amyloid beta (Aß) plaques in the brain. Aß1-42 is the main component of Aß plaque, which is toxic to neuronal cells. Si nanowires (Si NWs) have the advantages of small particle size, high specific surface area, and good biocompatibility, and have potential application prospects in suppressing Aß aggregation. In this study, we employed the vapor-liquid-solid (VLS) growth mechanism to grow Si NWs using Au nanoparticles as catalysts in a plasma-enhanced chemical vapor deposition (PECVD) system. Subsequently, these Si NWs were transferred to a phosphoric acid buffer solution (PBS). We found that Si NWs significantly reduced cell death in PC12 cells (rat adrenal pheochromocytoma cells) induced by Aß1-42 oligomers via double staining with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and fluorescein diacetate/propyl iodide (FDA/PI). Most importantly, pre-incubated Si NWs largely prevented Aß1-42 oligomer-induced PC12 cell death, suggesting that Si NWs exerts an anti-Aß neuroprotective effect by inhibiting Aß aggregation. The analysis of Fourier Transform Infrared (FTIR) results demonstrates that Si NWs reduce the toxicity of fibrils and oligomers by intervening in the formation of ß-sheet structures, thereby protecting the viability of nerve cells. Our findings suggest that Si NWs may be a potential therapeutic agent for AD by protecting neuronal cells from the toxicity of Aß1-42.


Asunto(s)
Péptidos beta-Amiloides , Nanocables , Fármacos Neuroprotectores , Silicio , Animales , Ratas , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/toxicidad , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/antagonistas & inhibidores , Supervivencia Celular/efectos de los fármacos , Nanocables/química , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/química , Células PC12 , Fragmentos de Péptidos/química , Fragmentos de Péptidos/toxicidad , Fragmentos de Péptidos/farmacología , Agregado de Proteínas/efectos de los fármacos , Silicio/química
19.
Environ Toxicol ; 39(9): 4267-4277, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38700421

RESUMEN

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.


Asunto(s)
Péptidos beta-Amiloides , Plomo , Microglía , Estrés Oxidativo , Especies Reactivas de Oxígeno , Microglía/efectos de los fármacos , Microglía/metabolismo , Péptidos beta-Amiloides/toxicidad , Péptidos beta-Amiloides/metabolismo , Animales , Plomo/toxicidad , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fragmentos de Péptidos/toxicidad , Fragmentos de Péptidos/metabolismo , Ciclooxigenasa 2/metabolismo , Ratones , Línea Celular , Daño del ADN/efectos de los fármacos , Enfermedades Neurodegenerativas/inducido químicamente
20.
J Alzheimers Dis ; 99(3): 911-925, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38728187

RESUMEN

Background: Neuroinflammation plays a crucial part in the initial onset and progression of Alzheimer's disease (AD). NLRP3 inflammasome was demonstrated to get involved in amyloid-ß (Aß)-induced neuroinflammation. However, the mechanism of Aß-triggered activation of NLRP3 inflammasome remains poorly understood. Objective: Based on our previous data, the study aimed to identify the downstream signals that bridge the activation of TLR4 and NLRP3 inflammasome associated with Aß. Methods: BV-2 cells were transfected with TLR4siRNA or pretreated with a CLI-095 or NSC23766, followed by Aß1-42 treatment. APP/PS1 mice were injected intraperitoneally with CLI-095 or NSC23766. NLRP3 inflammasome and microglia activation was detected with immunostaining and western blot. G-LISA and Rac1 pull-down activation test were performed to investigate the activation of Rac1. Real-time PCR and ELISA were used to detect the inflammatory cytokines. Aß plaques were assessed by western blotting and immunofluorescence staining. Morris water maze test was conducted to determine the spatial memory in mice. Results: Rac1 and NLRP3 inflammasome were activated by Aß in both in vitro and in vivo experiments. Inhibition of TLR4 reduced the activity of Rac1 and NLRP3 inflammasome induced by Aß1-42. Furthermore, inhibition of Rac1 blocked NLRP3 inflammasome activation mediated by TLR4. Blocking the pathway by CLI095 or NSC23766 suppressed Aß1-42-triggered activation of microglia, reduced the expression of pro-inflammatory mediators and ameliorated the cognition deficits in APP/PS1 mice. Conclusions: Our study demonstrated that TLR4/Rac1/NLRP3 pathway mediated Aß-induced neuroinflammation, which unveiled a novel pathway and key contributors underlying the pathogenic mechanism of Aß.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Ratones Transgénicos , Proteína con Dominio Pirina 3 de la Familia NLR , Enfermedades Neuroinflamatorias , Receptor Toll-Like 4 , Proteína de Unión al GTP rac1 , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Péptidos beta-Amiloides/metabolismo , Receptor Toll-Like 4/metabolismo , Enfermedad de Alzheimer/metabolismo , Ratones , Proteína de Unión al GTP rac1/metabolismo , Enfermedades Neuroinflamatorias/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Microglía/metabolismo , Microglía/efectos de los fármacos , Inflamasomas/metabolismo , Masculino , Fragmentos de Péptidos/toxicidad , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Neuropéptidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA