Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.748
Filter
1.
Molecules ; 29(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38731472

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides , Nanowires , Silicon , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Nanowires/chemistry , Animals , PC12 Cells , Rats , Silicon/chemistry , Peptide Fragments/chemistry , Peptide Fragments/toxicity , Peptide Fragments/pharmacology , Cell Survival/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Protein Aggregates/drug effects , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism
2.
J Neuroinflammation ; 21(1): 144, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822334

ABSTRACT

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.


Subject(s)
CD11b Antigen , Encephalomyelitis, Autoimmune, Experimental , Interferon-gamma , Mice, Inbred C57BL , Myeloid Cells , Spleen , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Interferon-gamma/metabolism , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/metabolism , Spleen/immunology , CD11b Antigen/metabolism , Female , Myelin-Oligodendrocyte Glycoprotein/toxicity , Myelin-Oligodendrocyte Glycoprotein/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Peptide Fragments/toxicity , Peptide Fragments/pharmacology , Transforming Growth Factor beta/metabolism , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/immunology , Forkhead Transcription Factors/metabolism , Disease Models, Animal
3.
J Alzheimers Dis ; 99(3): 911-925, 2024.
Article in English | MEDLINE | ID: mdl-38728187

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein , Neuroinflammatory Diseases , Toll-Like Receptor 4 , rac1 GTP-Binding Protein , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Amyloid beta-Peptides/metabolism , Toll-Like Receptor 4/metabolism , Alzheimer Disease/metabolism , Mice , rac1 GTP-Binding Protein/metabolism , Neuroinflammatory Diseases/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Microglia/metabolism , Microglia/drug effects , Inflammasomes/metabolism , Male , Peptide Fragments/toxicity , Mice, Inbred C57BL , Disease Models, Animal , Neuropeptides
4.
J Neuroimmune Pharmacol ; 19(1): 24, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38780885

ABSTRACT

Cornuside has been discovered to improve learning and memory in AD mice, however, its underlying mechanism was not fully understood. In the present study, we established an AD mice model by intracerebroventricular injection of Aß1-42, which were treated with cornuside (3, 10, 30 mg/kg) for 2 weeks. Cornuside significantly ameliorated cognitive function of AD mice in series of behavioral tests, including Morris water maze test, nest building test, novel object recognition test and step-down test. Additionally, cornuside could attenuate neuronal injury, and promote cholinergic synaptic transmission by restoring the level of acetylcholine (ACh) via inhibiting acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE), as well as facilitating choline acetyltransferase (ChAT). Furthermore, cornuside inhibited oxidative stress levels amplified as decreased malondialdehyde (MDA), by inhibiting TXNIP expression, improving total anti-oxidative capacity (TAOC), raising activities of superoxide dismutase (SOD) and catalase (CAT). Cornuside also reduced the activation of microglia and astrocytes, decreased the level of proinflammatory factors TNF-α, IL-6, IL-1ß, iNOS and COX2 via interfering RAGE-mediated IKK-IκB-NF-κB phosphorylation. Similar anti-oxidative and anti-inflammatory effects were also found in LPS-stimulated BV2 cells via hampering RAGE-mediated TXNIP activation and NF-κB nuclear translocation. Virtual docking revealed that cornuside could interact with the active pocket of RAGE V domain directly. In conclusion, cornuside could bind to the RAGE directly impeding the interaction of Aß and RAGE, and cut down the expression of TXNIP inhibiting ROS production and oxidative stress, as well as hamper NF-κB p65 mediated the inflammation.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Cognitive Dysfunction , NF-kappa B , Peptide Fragments , Receptor for Advanced Glycation End Products , Signal Transduction , Animals , Mice , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/chemically induced , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/metabolism , Peptide Fragments/toxicity , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/chemically induced , Signal Transduction/drug effects , Receptor for Advanced Glycation End Products/metabolism , NF-kappa B/metabolism , Male , Oxidative Stress/drug effects
5.
J Alzheimers Dis ; 99(2): 477-483, 2024.
Article in English | MEDLINE | ID: mdl-38669543

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disease with limited therapeutic strategies. NB-02 is a novel botanical drug that has shown promise as a protective and therapeutic treatment for AD in an APP/PS1 preclinical mouse model. In this paper, we investigate the underlying mechanisms by which NB-02 provides these therapeutic advantages using in vitro neuron-astrocyte co-cultures. Pretreatment with NB-02 prevented pathological calcium elevations in neurons and astrocytes after application of toxic soluble amyloid-ß (Aß) oligomers. NB-02 also prevented cell death associated with the addition of soluble Aß oligomers suggesting NB-02 is effective at protecting both neurons and astrocytes from Aß-mediated damage.


Subject(s)
Amyloid beta-Peptides , Astrocytes , Coculture Techniques , Neurons , Neuroprotective Agents , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Mice , Cells, Cultured , Calcium/metabolism , Peptide Fragments/toxicity , Peptide Fragments/pharmacology , Humans
6.
J Toxicol Environ Health A ; 87(10): 436-447, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38557424

ABSTRACT

One of the main pathological features noted in Alzheimer's disease (AD) is the presence of plagues of aggregated ß-amyloid (Aß1-42)-peptides. Excess deposition of amyloid-ß oligomers (AßO) are known to promote neuroinflammation. Sequentially, following neuroinflammation astrocytes become activated with cellular characteristics to initiate activated astrocytes. The purpose of this study was to determine whether total flavonoids derived from Dracocephalum moldavica L. (TFDM) inhibited Aß1-42-induced damage attributed to activated C8-D1A astrocytes. Western blotting and ELISA were used to determine the expression of glial fibrillary acidic protein (GFAP), and complement C3 to establish the activation status of astrocytes following induction from exposure to Aß1-42. Data demonstrated that stimulation of C8-D1A astrocytes by treatment with 40 µM Aß1-42 for 24 hr produced significant elevation in protein expression and protein levels of acidic protein (GFAP) and complement C3 accompanied by increased expression and levels of inflammatory cytokines. Treatment with TFDM or the clinically employed drug donepezil in AD therapy reduced production of inflammatory cytokines, and toxicity initiated following activation of C8-D1A astrocytes following exposure to Aß1-42. Therefore, TFDM similar to donepezil inhibited inflammatory secretion in reactive astrocytes, suggesting that TFDM may be considered as a potential compound to be utilized in AD therapy.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Lamiaceae , Humans , Amyloid beta-Peptides/pharmacology , Alzheimer Disease/drug therapy , Flavonoids/pharmacology , Complement C3/metabolism , Complement C3/pharmacology , Complement C3/therapeutic use , Neuroinflammatory Diseases , Astrocytes/metabolism , Donepezil/metabolism , Donepezil/pharmacology , Donepezil/therapeutic use , Cytokines/metabolism , Peptide Fragments/metabolism , Peptide Fragments/toxicity
7.
Neuroscience ; 547: 37-55, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38604526

ABSTRACT

The Aß hypothesis has long been central to Alzheimer's disease (AD) theory, with a recent surge in attention following drug approvals targeting Aß plaque clearance. Aß42 oligomers (AßO) are key neurotoxins. While ß-amyloid (Aß) buildup is a hallmark of AD, postmortem brain analyses have unveiled human islet amyloid polypeptide (hIAPP) deposition in AD patients, suggesting a potential role in Alzheimer's pathology. This study investigates the neurotoxic effects of co-aggregates of Aß42 and hIAPP, specifically focusing on their impact on cell survival, apoptosis, and AD-like pathology. We analyzed and compared the impact of AßO and Aß42-hIAPP on cell survival in SH-SY5Y cells, apoptosis and inducing AD-like pathology in glutamatergic neurons. Aß42-hIAPP co-oligomers exhibited significantly greater toxicity, causing 2.3-3.5 times higher cell death compared to AßO alone. Furthermore, apoptosis rates were significantly exacerbated in glutamatergic neurons when exposed to Aß42-hIAPP co-oligomers. The study also revealed that Aß42-hIAPP co-oligomers induced typical AD-like pathology in glutamatergic neurons, including the presence of Aß deposits (detected by 6E10 and 4G8 immunofluorescence) and alterations in tau protein (changes in total tau HT7, phosphorylated tau AT8, AT180). Notably, Aß42-hIAPP co-oligomers induced a more severe AD pathology compared to AßO alone. These findings provide compelling evidence for the heightened toxicity of Aß42-hIAPP co-oligomers on neurons and their role in exacerbating AD pathology. The study contributes novel insights into the pathogenesis of Alzheimer's disease, highlighting the potential involvement of hIAPP in AD pathology. Together, these findings offer novel insights into AD pathogenesis and routes for constructing animal models.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Apoptosis , Cell Survival , Islet Amyloid Polypeptide , Neurons , Peptide Fragments , Humans , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Peptide Fragments/toxicity , Peptide Fragments/metabolism , Neurons/metabolism , Neurons/pathology , Neurons/drug effects , Islet Amyloid Polypeptide/metabolism , Islet Amyloid Polypeptide/toxicity , Apoptosis/drug effects , Apoptosis/physiology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , tau Proteins/metabolism
8.
Neurosci Lett ; 825: 137708, 2024 Mar 10.
Article in English | MEDLINE | ID: mdl-38438068

ABSTRACT

Dehydroeffusol, a major phenanthrene in Juncus effusus, protects neurodegeneration induced by intracellular Zn2+ ferried by extracellular amyloid ß1-42 (Aß1-42). Here we focused on adrenaline ß receptor activation and the induction of metallothioneins (MTs), intracellular Zn2+-binding proteins to test the protective mechanism of dehydroeffusol. Isoproterenol, an agonist of adrenergic ß receptors elevated the level of MTs in the dentate granule cell layer 1 day after intracerebroventricular (ICV) injection. When Aß1-42 was injected 1 day after isoproterenol injection, pre-injection of isoproterenol protected Aß1-42 toxicity via reducing the increase in intracellular Zn2+ after ICV injection of Aß1-42. On the basis of the effect of increased MTs by isoproterenol, dehydroeffusol (15 mg/kg body weight) was orally administered to mice once a day for 2 days. On day later, dehydroeffusol elevated the level of MTs and prevented Aß1-42 toxicity via reducing Aß1-42-mediated increase in intracellular Zn2+. In contrast, propranolol, an antagonist of adrenergic ß receptors reduced the level of MTs increased by dehydroeffusol, resulting in invalidating the preventive effect of dehydroeffusol on Aß1-42 toxicity. The present study indicates that blockage of MT synthesis via adrenaline ß receptor activation invalidates dehydroeffusol-mediated prevention of Aß1-42 toxicity. It is likely that MT synthesis via adrenaline ß receptor activation is beneficial to neuroprotection and that oral intake of dehydroeffusol preventively serves against the Aß1-42 toxicity.


Subject(s)
Amyloid beta-Peptides , Metallothionein , Phenanthrenes , Mice , Animals , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/metabolism , Epinephrine , Isoproterenol , Receptors, Adrenergic, beta , Peptide Fragments/toxicity , Peptide Fragments/metabolism
9.
Exp Eye Res ; 242: 109861, 2024 May.
Article in English | MEDLINE | ID: mdl-38522635

ABSTRACT

Amyloid-beta (Aß), a family of aggregation-prone and neurotoxic peptides, has been implicated in the pathophysiology of age-related macular degeneration (AMD). We have previously shown that oligomeric and fibrillar species of Aß42 exerted retinal toxicity in rats, but while the consequences of exposure to amyloid were related to intracellular effects, the mechanism of Aß42 internalization in the retina is not well characterized. In the brain, the 67 kDa laminin receptor (67LR) participates in Aß-related neuronal cell death. A short peptide derived from pigment epithelium-derived factor (PEDF), formerly designated PEDF-335, was found to mitigate experimental models of ischemic retinopathy via targeting of 67LR. In the present study, we hypothesized that 67LR mediates the uptake of pathogenic Aß42 assemblies in the retina, and that targeting of this receptor by PEDF-335 may limit the internalization of Aß, thereby ameliorating its retinotoxicity. To test this assumption ARPE-19 cells in culture were incubated with PEDF-335 before treatment with fibrillar or oligomeric structures of Aß42. Immunostaining confirmed that PEDF-335 treatment substantially prevented amyloid internalization into ARPE-19 cells and maintained their viability in the presence of toxic oligomeric and fibrillar Aß42 entities in vitro. FRET competition assay was performed and confirmed the binding of PEDF-335 to 67LR in RPE-like cells. Wild-type rats were treated with intravitreal PEDF-335 in the experimental eye 2 days prior to administration of retinotoxic Aß42 oligomers or fibrils to both eyes. Retinal function was assessed by electroretinography through 6 weeks post injection. The ERG responses in rats treated with oligomeric or fibrillar Aß42 assemblies were near-normal in eyes previously treated with intravitreal PEDF-335, whereas those measured in the control eyes treated with injection of the Aß42 assemblies alone showed pathologic attenuation of the retinal function through 6 weeks. The retinal presence of 67LR was determined ex vivo by immunostaining and western blotting. Retinal staining demonstrated the constitutional expression of 67LR mainly in the retinal nuclear layers. In the presence of Aß42, the levels of 67LR were increased, although its retinal distribution remained largely unaltered. In contrast, no apparent differences in the retinal expression level of 67LR were noted following exposure to PEDF-335 alone, and its pattern of localization in the retina remained similarly concentrated primarily in the inner and outer nuclear layers. In summary, we found that PEDF-335 confers protection against Aß42-mediated retinal toxicity, with significant effects noted in cells as well as in vivo in rats. The effects of PEDF-335 in the retina are potentially mediated via binding to 67LR and by at least partial inhibition of Aß42 internalization. These results suggest that PEDF-335 may merit further consideration in the development of targeted inhibition of amyloid-related toxicity in the retina. More broadly, our observations provide evidence on the importance of extracellular versus intracellular Aß42 in the retina and suggest concepts on the molecular mechanism of Aß retinal pathogenicity.


Subject(s)
Amyloid beta-Peptides , Electroretinography , Eye Proteins , Nerve Growth Factors , Serpins , Animals , Serpins/metabolism , Eye Proteins/metabolism , Nerve Growth Factors/metabolism , Rats , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/metabolism , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Peptide Fragments/toxicity , Disease Models, Animal , Receptors, Laminin/metabolism , Male , Retina/drug effects , Retina/metabolism , Humans , Intravitreal Injections , Blotting, Western , Retinal Diseases/prevention & control , Retinal Diseases/metabolism , Retinal Diseases/chemically induced , Cells, Cultured
10.
Chem Biol Interact ; 394: 110978, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38552766

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by the accumulation of amyloid-ß (Aß) protein aggregates, leading to synaptic dysfunction and neuronal cell death. In this study, we used a comprehensive approach encompassing in vitro assays, computational analyses, and an in vivo Caenorhabditis elegans model to evaluate the inhibitory effects of various xanthones, focusing on Garcinone D (GD), on Aß42 oligomer formation. Dot blot analysis revealed concentration-dependent responses among xanthones, with GD consistently inhibiting Aß42 oligomer formation at low concentrations (0.1 and 0.5 µM, inhibitions of 84.66 ± 2.25% and 85.06 ± 6.57%, respectively). Molecular docking and dynamics simulations provided insights into the molecular interactions between xanthones and Aß42, highlighting the disruption of key residues involved in Aß42 aggregation. The neuroprotective potential of GD was established using transgenic C. elegans GMC101, with substantial delays in paralysis reported at higher concentrations. Our findings show that GD is a potent suppressor of Aß42 oligomer formation, suggesting its potential as a therapeutic candidate for AD. The concentration-dependent effects observed in both in vitro and in vivo models underscore the need for nuanced dose-response assessments. These findings contribute novel insights into the therapeutic landscape of xanthones against AD, emphasizing the multifaceted potential of GD for further translational endeavors in neurodegenerative disorder research.


Subject(s)
Amyloid beta-Peptides , Caenorhabditis elegans , Peptide Fragments , Xanthones , Animals , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Animals, Genetically Modified , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/metabolism , Disease Models, Animal , Molecular Docking Simulation , Molecular Dynamics Simulation , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Peptide Fragments/toxicity , Peptide Fragments/metabolism , Protein Aggregates/drug effects , Xanthones/pharmacology , Xanthones/chemistry
11.
J Alzheimers Dis ; 98(1): 163-186, 2024.
Article in English | MEDLINE | ID: mdl-38393907

ABSTRACT

Background: Increased blood-brain barrier (BBB) permeability and amyloid-ß (Aß) peptides (especially Aß1-42) (Aß42) have been linked to Alzheimer's disease (AD) pathogenesis, but the nature of their involvement in AD-related neuropathological changes leading to cognitive changes remains poorly understood. Objective: To test the hypothesis that chronic extravasation of bloodborne Aß42 peptide and brain-reactive autoantibodies and their entry into the brain parenchyma via a permeable BBB contribute to AD-related pathological changes and cognitive changes in a mouse model. Methods: The BBB was rendered chronically permeable through repeated injections of Pertussis toxin (PT), and soluble monomeric, fluorescein isothiocyanate (FITC)-labeled or unlabeled Aß42 was injected into the tail-vein of 10-month-old male CD1 mice at designated intervals spanning ∼3 months. Acquisition of learned behaviors and long-term retention were assessed via a battery of cognitive and behavioral tests and linked to neuropathological changes. Results: Mice injected with both PT and Aß42 demonstrated a preferential deficit in the capacity for long-term retention and an increased susceptibility to interference in selective attention compared to mice exposed to PT or saline only. Immunohistochemical analyses revealed increased BBB permeability and entry of bloodborne Aß42 and immunoglobulin G (IgG) into the brain parenchyma, selective neuronal binding of IgG and neuronal accumulation of Aß42 in animals injected with both PT and Aß42 compared to controls. Conclusion: Results highlight the potential synergistic role of BBB compromise and the influx of bloodborne Aß42 into the brain in both the initiation and progression of neuropathologic and cognitive changes associated with AD.


Subject(s)
Alzheimer Disease , Blood-Brain Barrier , Male , Mice , Animals , Blood-Brain Barrier/metabolism , Alzheimer Disease/pathology , Peptide Fragments/toxicity , Peptide Fragments/metabolism , Brain/pathology , Amyloid beta-Peptides/metabolism , Cognition , Immunoglobulin G/metabolism
12.
J Chem Neuroanat ; 137: 102404, 2024 04.
Article in English | MEDLINE | ID: mdl-38423257

ABSTRACT

Alzheimer's disease (AD) is a degenerative neurological disorder with unclear pathogenesis. Single-target drugs have very limited efficacy in treating AD, but synthetic multi-target drugs have poor efficacy and safety. Therefore, finding suitable natural multi-target drugs against AD is of great interest for research studies. We chose two flavonols, myricetin and morin, for the relevant study. In this study, we used microinjection of Aß1-42 oligomers into the CA1 region of rat hippocampus, combined with gavage of Aluminum chloride hexahydrate (AlCl3·6H2O) solution to establish AD rat models, and myricetin and morin were selected as intervening drugs to explore the protective effects against neurological impairment. Experimental results showed that myricetin or morin could reduce the production of Aß, Tubulin-associated unit (Tau), and Phosphorylated tubulin-associated unit (p-Tau), down-regulate the expression of relevant inflammatory factors, reduce hippocampal cell apoptosis in rats. There was a significant increase in the activity of adenosine triphosphatase, catalase, total superoxide dismutase, and the content of glutathione in the brain tissue. However, the content of malondialdehyde, inducible nitric oxide synthase, and the activity of acetylcholinesterase were decreased in the brain tissue. These two flavonols can regulate the imbalance of monoamine and amino acid neurotransmitter levels. In conclusion, Myricetin or morin can effectively improve learning and memory dysfunction in AD rats induced by Aß1-42/Al3+ through anti-oxidative stress and anti-apoptotic features.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Disease Models, Animal , Flavones , Flavonoids , Neuroprotective Agents , Peptide Fragments , Animals , Flavonoids/pharmacology , Flavonoids/therapeutic use , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/chemically induced , Alzheimer Disease/pathology , Rats , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Peptide Fragments/toxicity , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Male , Rats, Sprague-Dawley , Aluminum Chloride/toxicity , Hippocampus/drug effects , Hippocampus/metabolism , Oxidative Stress/drug effects
13.
J Hazard Mater ; 465: 133518, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38228001

ABSTRACT

Nanoplastics, widely existing in the environment and organisms, have been proven to cross the blood-brain barrier, increasing the incidence of neurodegenerative diseases like Alzheimer's disease (AD). However, current studies mainly focus on the neurotoxicity of nanoplastics themselves, neglecting their synergistic effects with other biomolecules and the resulting neurotoxicity. Amyloid ß peptide (Aß), which triggers neurotoxicity through its self-aggregation, is the paramount pathogenic protein in AD. Here, employing polystyrene nanoparticles (PS) as a model for nanoplastics, we reveal that 100 pM PS nanoparticles significantly accelerate the nucleation rate of two Aß subtypes (Aß40 and Aß42) at low concentrations, promoting the formation of more Aß oligomers and leading to evident neurotoxicity. The hydrophobic surface of PS facilitates the interaction of hydrophobic fragments between Aß monomers, responsible for the augmented neurotoxicity. This work provides consequential insights into the modulatory impact of low-dose PS on Aß aggregation and the ensuing neurotoxicity, presenting a valuable foundation for future research on the intricate interplay between environmental toxins and brain diseases.


Subject(s)
Alzheimer Disease , Nanoparticles , Humans , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Microplastics , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/toxicity
14.
Alzheimers Res Ther ; 16(1): 13, 2024 01 18.
Article in English | MEDLINE | ID: mdl-38238842

ABSTRACT

BACKGROUND: Amyloid-ß42 (Aß42) aggregation consists of a complex chain of nucleation events producing soluble oligomeric intermediates, which are considered the major neurotoxic agents in Alzheimer's disease (AD). Cerebral lesions in the brain of AD patients start to develop 20 years before symptom onset; however, no preventive strategies, effective treatments, or specific and sensitive diagnostic tests to identify people with early-stage AD are currently available. In addition, the isolation and characterisation of neurotoxic Aß42 oligomers are particularly difficult because of their transient and heterogeneous nature. To overcome this challenge, a rationally designed method generated a single-domain antibody (sdAb), named DesAb-O, targeting Aß42 oligomers. METHODS: We investigated the ability of DesAb-O to selectively detect preformed Aß42 oligomers both in vitro and in cultured neuronal cells, by using dot-blot, ELISA immunoassay and super-resolution STED microscopy, and to counteract the toxicity induced by the oligomers, monitoring their interaction with neuronal membrane and the resulting mitochondrial impairment. We then applied this approach to CSF samples (CSFs) from AD patients as compared to age-matched control subjects. RESULTS: DesAb-O was found to selectively detect synthetic Aß42 oligomers both in vitro and in cultured cells, and to neutralise their associated neuronal dysfunction. DesAb-O can also identify Aß42 oligomers present in the CSFs of AD patients with respect to healthy individuals, and completely prevent cell dysfunction induced by the administration of CSFs to neuronal cells. CONCLUSIONS: Taken together, our data indicate a promising method for the improvement of an early diagnosis of AD and for the generation of novel therapeutic approaches based on sdAbs for the treatment of AD and other devastating neurodegenerative conditions.


Subject(s)
Alzheimer Disease , Single-Domain Antibodies , Humans , Alzheimer Disease/pathology , Single-Domain Antibodies/therapeutic use , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/metabolism , Enzyme-Linked Immunosorbent Assay , Brain/metabolism , Peptide Fragments/toxicity
15.
Folia Neuropathol ; 62(1): 47-58, 2024.
Article in English | MEDLINE | ID: mdl-38174688

ABSTRACT

Alzheimer's disease (AD), the most common contributor to dementia, is a growing global health problem. This study aimed to investigate the role of lemur tyrosine kinase 2 (LMTK2) in AD as well as its relevant mechanism. To establish an in vitro cell model, PC12 cells were challenged with 20 µmol/l Ab 25-35 for 24 h. RT-qPCR and western blot examined LMTK2 mRNA and protein expressions. With the application of CCK-8, TUNEL, iron colorimetric assay kit and DCFH-DA, the viability, apoptosis, Fe 2+ and ROS content in PC12 cells were assessed. Besides, the expressions of oxidative stress-, apoptosis-, ferroptosis- and Nrf2/ARE signalling-related proteins were evaluated with western blot. Moreover, commercial kits examined SOD, MDA and CAT contents. The results manifested that LMTK2 expression was noticeably downregulated in Ab 25-35 -treated PC12 cells. Notably, LMTK2 overexpression exhibited inhibitory effects on oxidative stress, apoptosis and ferroptosis in PC12 cells exposed to Ab 25-35 . The upregulated Nrf2, NQO1 and HO-1 expressions in LMTK2 overexpressed-PC12 cells with Ab 25-35 induction revealed that LMTK2 overexpression could activate the Nrf2/ARE signalling pathway. What is more, a series of cellular experiments further testified that ML385, a specific Nrf2 inhibitor, partly hindered the protective role of LMTK2 overexpression against Ab 25-35 -triggered oxidative stress, apoptosis and ferroptosis in PC12 cells. In conclusion, LMTK2 overexpression alleviated the ferroptosis, oxidant damage and apoptosis in PC12 cells exposed to Ab 25-35 through the activation of the Nrf2/ARE signalling pathway, indicating the potential target of LMTK2 in the treatment of AD.


Subject(s)
Amyloid beta-Peptides , Apoptosis , Ferroptosis , NF-E2-Related Factor 2 , Oxidative Stress , Peptide Fragments , Signal Transduction , PC12 Cells , Animals , Oxidative Stress/drug effects , Rats , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Apoptosis/drug effects , Ferroptosis/drug effects , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/metabolism , Peptide Fragments/toxicity , Alzheimer Disease/metabolism , Alzheimer Disease/pathology
16.
Neurochem Res ; 49(1): 99-116, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37615884

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disease that is the main form of dementia. Abnormal deposition of amyloid-beta (Aß) peptides in neurons and synapses cause neuronal loss and cognitive deficits. We have previously reported that ferroptosis and necroptosis were implicated in Aß25-35 neurotoxicity, and their specific inhibitors had attenuating effects on cognitive impairment induced by Aß25-35 neurotoxicity. Here, we aimed to examine the impact of ferroptosis and necroptosis inhibition following the Aß25-35 neurotoxicity on the neuronal excitability of dentate gyrus (DG) and the possible involvement of voltage-gated Ca2+ channels in their effects. After inducing Aß25-35 neurotoxicity, electrophysiological alterations in the intrinsic properties and excitability were recorded by the whole-cell patch-clamp under current-clamp condition. Voltage-clamp recordings were also performed to shed light on the involvement of calcium channel currents. Aß25-35 neurotoxicity induced a considerable reduction in input resistance (Rin), accompanied by a profoundly decreased excitability and a reduction in the amplitude of voltage-gated calcium channel currents in the DG granule cells. However, three days of administration of either ferrostatin-1 (Fer-1), a ferroptosis inhibitor, or Necrostatin-1 (Nec-1), a necroptosis inhibitor, in the entorhinal cortex could almost preserve the normal excitability and the Ca2+ currents. In conclusion, these findings suggest that ferroptosis and necroptosis involvement in EC amyloidopathy could be a potential candidate to prevent the suppressive effect of Aß on the Ca2+ channel current and neuronal function, which might take place in neurons during the development of AD.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Neuroprotective Agents , Humans , Neuroprotective Agents/pharmacology , Peptide Fragments/toxicity , Peptide Fragments/metabolism , Amyloid beta-Peptides/metabolism , Calcium Channels , Dentate Gyrus
17.
Inflammopharmacology ; 32(2): 1461-1474, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37758932

ABSTRACT

Lately, interest surrounding the utilization of plant-derived compounds as a viable beneficial approach for treating Alzheimer's disease (AD) has significantly increased. This study aimed to assess the defensive properties of rosavin against Alzheimer's disease induced by amyloid-ß, utilizing experimental models. We found that rosavin exhibited anti-aggregation and disaggregation properties, suggesting its potential to prevent the gathering of Aß-aggregates. In vitro experiments revealed that rosavin effectively mitigated the neurotoxicity induced by Aß in Neuro-2a cells, showcasing its protective potential. Rosavin significantly improved the Aß-induced cognitive deficits in Wistar rats, particularly in spatial memory. Which the pathophysiology of AD includes oxidative damage, which negatively impacts biological macromolecules. Triggers the apoptotic process, causing macromolecular destruction. Interestingly, rosavin attenuated Aß-induced macromolecular damages, thereby preserving neuronal integrity. Furthermore, the activation of antioxidative defense enzymes by rosavin inhibited oxidative damage. The positive outcomes associated with rosavin were primarily attributed to its capacity to enhance acetylcholine-mediated effects. Finally, rosavin has the potential to alleviate Aß-induced neurotoxicity and macromolecular damages, ultimately resulting in enhanced memorial and reasoning function in Wistar rats, offering promising prospects for the treatment of AD.


Subject(s)
Alzheimer Disease , Rats , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/chemically induced , Rats, Wistar , Amyloid beta-Peptides/toxicity , Disaccharides/adverse effects , Peptide Fragments/toxicity , Disease Models, Animal
18.
Neurosci Lett ; 818: 137564, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38013121

ABSTRACT

OBJECTIVE: ß-Amyloid (Aß) induced neurotoxicity is an implicated mechanism in Alzheimer's disease (AD). This study focused on the role of GDP dissociation inhibitor 1 (GDI1) in Aß-induced neurotoxicity. METHODS: Data from the GEO database for AD-related datasets GSE140829, GSE63061, GSE36980, and GSE60360 were downloaded and identified common differentially expressed genes (coDEGs). The mRNA levels of GDI1 in the serum of AD patients were analyzed by RT-qPCR. ROC curve evaluated the diagnostic value. Aß25-35 induced SH-SY5Y cells to construct an AD cell model. CCK-8, flow cytometry, and ELISA assay were used to analyze cell viability, apoptosis, and concentrations of inflammatory factors. KEGG enrichment was employed to analyze the signal pathway of targets from GDI1 in the AD. RESULTS: The GEO database identifies coDEGs including GDI1. GDI1 is generally elevated in serum from AD patients as well as in Aß-induced SH-SY5Y cells. GDI1 has 77.97% sensitivity and 84.44% specificity to identify AD patients from controls. Aß induced decreased cell viability, increased apoptosis, and promoted over-secretion of inflammatory cytokines, but they were all partially weakened by reduced GDI1. What's more, the GDI1 interacting gene and AD target gene were co-enriched on Endocytosis and MAPK signaling pathway. CONCLUSION: Elevated GDI1 is a potential diagnostic biomarker for AD and that inhibition of GDI1 attenuates Aß-induced neurotoxicity in AD. Our study offers new horizons for AD treatment.


Subject(s)
Alzheimer Disease , Neuroblastoma , Humans , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apoptosis , Cell Line, Tumor , Guanine Nucleotide Dissociation Inhibitors , Peptide Fragments/toxicity , Peptide Fragments/metabolism
19.
Bioorg Med Chem ; 96: 117536, 2023 12 15.
Article in English | MEDLINE | ID: mdl-38016411

ABSTRACT

In the present study, we evaluated the neuroprotective potential of Hesperidin Methyl Chalcone (HMC) against the neurotoxicity induced by Aß(25-35) peptide. HMC demonstrated higher free-radical scavenging activity than Hesperidin in initial cell-free studies. Investigations using the fluorescent dye thioflavin T with Aß(25-35) peptide showed that HMC has the ability to combat extracellular amyloid aggregation by possessing anti-aggregation property against oligomers and by disaggregating mature fibrils. Also, the results of the molecular simulation studies show that HMC ameliorated oligomer formation. Further, the anti-Alzheimer's property of HMC was investigated in in vitro cell conditions by pre-treating the neuro 2a (N2a) cells with HMC before inducing Aß(25-35) toxicity. The findings demonstrate that HMC increased cell viability, reduced oxidative stress, prevented macromolecular damage, allayed mitochondrial dysfunction, and exhibited anticholinesterase activity. HMC also reduced Aß induced neuronal cell death by modulating caspase-3 activity, Bax expression and Bcl2 overexpression, demonstrating that HMC pre-treatment reduced mitochondrial damage and intrinsic apoptosis induced by Aß(25-35).In silico evaluation against potential AD targets reveal that HMC could be a potent inhibitor of BACE-1, inhibiting the formation of toxic Aß peptides. Overall, the findings imply that the neuroprotective efficacy of HMC has high prospects for addressing a variety of pathogenic consequences caused by amyloid beta in AD situations and alleviating cognitive impairments.


Subject(s)
Alzheimer Disease , Chalcones , Hesperidin , Neuroprotective Agents , Humans , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/metabolism , Chalcones/pharmacology , Hesperidin/pharmacology , Amyloid , Peptide Fragments/toxicity , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology
20.
J Mol Neurosci ; 73(11-12): 932-945, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37882913

ABSTRACT

Alzheimer's disease (AD) is a prevalently neurodegenerative disease characterized by neuronal damage which is associated with amyloid-ß (Aß) accumulation. Hederagenin is a triterpenoid saponin, exerting anti-apoptotic, anti-oxidative, anti-inflammatory, anti-tumoral, and neuroprotective activities. However, its role in AD progression is still obscure. The aim of this study was to explore the influences of hederagenin on Aß-caused neuronal injury in vitro. Neuronal cells were treated with Aß25-35 (Aß) to establish a cellular model of AD. Cell viability was assessed using cell counting kit-8 (CCK-8). Oxidative stress was evaluated by detecting reactive oxygen species (ROS) generation and superoxide dismutase (SOD) activity. Apoptosis was investigated using TUNEL staining and caspase-3 activity assays. Protein tyrosine phosphatase nonreceptor type 1 (PTPN1) was screened by bioinformatics analysis. Protein levels of PTPN1 and protein kinase B (Akt) were measured by western blotting. Hederagenin (2.5, 5, and 10 µM) alone did not affect viability of neuronal cells, but relieved Aß-induced viability reduction. Hederagenin mitigated Aß-induced increase in ROS accumulation and decrease in SOD activity. Hederagenin attenuated Aß-induced increase in apoptotic rate and caspase-3 activity. PTPN1 was screened as a target of hederagenin against AD by bioinformatics analysis. Hederagenin treatment resisted Aß-induced decrease in PTPN1 mRNA and protein levels in neuronal cells. PTPN1 silencing attenuated the suppressive functions of hederagenin in Aß-stimulated oxidative stress and apoptosis. Hederagenin mitigated Aß-induced Akt signaling inactivation by upregulating PTPN1 expression. In conclusion, hederagenin attenuates oxidative stress and apoptosis in neuronal cells stimulated with Aß by promoting PTPN1/Akt signaling activation.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Neuroprotective Agents , Humans , Reactive Oxygen Species/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Neurodegenerative Diseases/drug therapy , Phosphoric Monoester Hydrolases , Caspase 3/metabolism , Oxidative Stress , Amyloid beta-Peptides/toxicity , Amyloid beta-Peptides/metabolism , Alzheimer Disease/metabolism , Apoptosis , Superoxide Dismutase-1/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Peptide Fragments/toxicity , Peptide Fragments/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...