Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Nutr Neurosci ; 25(10): 2167-2180, 2022 Oct.
Article in English | MEDLINE | ID: mdl-33993853

ABSTRACT

Fucoxanthin, one of the most abundant carotenoids from edible brown seaweeds, for years has been used as a bioactive dietary supplement and functional food ingredient. Recently, fucoxanthin was reported to penetrate the blood-brain barrier, and was superior to other carotenoids to exert anti-neurodegenerative disorder effects via acting on multiple targets, including amyloid protein aggregation, oxidative stress, neuroinflammation, neuronal loss, neurotransmission dysregulation and gut microbiota disorder. However, the concentration of fucoxanthin required for in vivo neuroprotective effects is somewhat high, and the poor bioavailability of this molecule might prevent its clinical use. As such, new strategies have been introduced to overcome these obstacles, and may help to develop fucoxanthin as a novel lead for neurodegenerative disorders. Moreover, it has been shown that some metabolites of fucoxanthin may produce potent in vivo neuroprotective effects. Altogether, these studies suggest the possibility for future development of fucoxanthin as a one-compound-multiple-target or pro-drug type pharmaceutical or nutraceutical treatment for neurodegenerative disorders.Trial registration: ClinicalTrials.gov identifier: NCT03625284.Trial registration: ClinicalTrials.gov identifier: NCT02875392.Trial registration: ClinicalTrials.gov identifier: NCT03613740.Trial registration: ClinicalTrials.gov identifier: NCT04761406.


Subject(s)
Food Ingredients , Neurodegenerative Diseases , Neuroprotective Agents , Prodrugs , Carotenoids , Clinical Trials as Topic , Humans , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Protein Aggregates , Xanthophylls
2.
ACS Chem Neurosci ; 12(13): 2462-2477, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34156230

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder with multiple pathological features. Therefore, a multitarget-directed ligands (MTDLs) strategy has been developed to treat AD. We have previously designed and synthesized dimeric tacrine(10)-hupyridone (A10E), a novel tacrine derivative with acetylcholinesterase (AChE) inhibition and brain-derived neurotrophic factor (BDNF) activation activity, by linking tacrine and a fragment of huperzine A. However, it was largely unknown whether A10E could act on other AD targets and produce cognitive-enhancing ability in AD animal models. In this study, A10E could prevent cognitive impairments in APP/PS1 transgenic mice and ß-amyloid (Aß) oligomers-treated mice, with higher potency than tacrine and huperzine A. Moreover, A10E could effectively inhibit Aß production and deposition, alleviate neuroinflammation, enhance BDNF expression, and elevate cholinergic neurotransmission in vivo. At nanomolar concentrations, A10E could inhibit Aß oligomers-induced neurotoxicity via the activation of tyrosine kinase receptor B (TrkB)/Akt pathway in SH-SY5Y cells. Furthermore, Aß oligomerization and fibrillization could be directly disrupted by A10E. Importantly, A10E at high concentrations did not produce obvious hepatotoxicity. Our results indicated that A10E could produce anti-AD neuroprotective effects via the inhibition of Aß aggregation, the activation of the BDNF/TrkB pathway, the alleviation of neuroinflammation, and the decrease of AChE activity. As MTDLs could produce additional benefits, such as overcoming the deficits of drug combination and enhancing the compliance of AD patients, our results also suggested that A10E might be developed as a promising MTDL lead for the treatment of AD.


Subject(s)
Alzheimer Disease , Tacrine , Alzheimer Disease/drug therapy , Amyloid beta-Peptides , Animals , Cholinesterase Inhibitors/pharmacology , Humans , Ligands , Mice , Tacrine/pharmacology
3.
Int Immunopharmacol ; 97: 107656, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33895476

ABSTRACT

OBJECTIVES: This study was aimed to investigate the neuroprotective effects of 9-methylfascaplysin, a novel marine derivative derived from sponge, against middle cerebral artery occlusion/reperfusion (MCAO)-induced motor impairments, neuroinflammation and oxidative stress in rats. METHODS: Neurological and behavioral tests were used to evaluate behavioral changes. The 2, 3, 5-triphenyltetrazolium chloride staining was used to determine infarct size and edema extent. Activated microglia/macrophage was analyzed by immunohistochemical staining of Iba-1. RT-PCR and ELISA were used to measure the expression of inducible nitric oxide synthase, tumor necrosis factor-α, interleukin-1ß, CD16 and CD206. Western blotting analysis was performed to explore the activation of nuclear factor-κB (NF-κB) and NLRP3. The levels of oxidative stress were studied by evaluating the activities of superoxide dismutase, catalase and glutathione peroxidase. RESULTS: Post-occlusion intracerebroventricular injection of 9-methylfascaplysin significantly attenuated motor impairments and infarct size in MCAO rats. Moreover, 9-methylfascaplysin reduced the activation of microglia/macrophage in ischemic penumbra as evidenced by the decreased Iba-1-positive area and the reduced expression of pro-inflammatory factors. Furthermore, 9-methylfascaplysin inhibited MCAO-induced oxidative stress and activation of NF-κB and NLRP3 inflammasome. CONCLUSION: All the results suggested that 9-methylfascaplysin might produce neuroprotective effects against MCAO via the reduction of oxidative stress and neuroinflammation, simultaneously, possibly via the inhibition of NF-κB and NLRP3 inflammasome.


Subject(s)
Indoles/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Neuroinflammatory Diseases/drug therapy , Neuroprotective Agents/pharmacology , Animals , Disease Models, Animal , Humans , Indoles/therapeutic use , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/pathology , Inflammasomes/antagonists & inhibitors , Inflammasomes/metabolism , Male , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/pathology , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Oxidative Stress/immunology , Rats , Signal Transduction/drug effects , Signal Transduction/immunology
4.
Int J Biol Macromol ; 170: 33-41, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33352157

ABSTRACT

Fast green FCF (FGF) is often used in foods, pharmaceuticals, and cosmetics. However, little is known about the interactions of FGF with amyloid-ß protein (Aß) associated with Alzheimer's disease. In this study, the inhibitory effects of FGF on Aß fibrillogenesis, the disruption of preformed Aß fibrils, the reduction of Aß-induced cytotoxicity, and the attenuation of Aß-induced learning and memory impairments in mice were investigated. FGF significantly inhibited Aß fibrillogenesis and disintegrated the mature fibrils as evidenced by thioflavin T fluorescence and atomic force microscopy studies. Co-incubation of Aß with FGF greatly reduced Aß-induced cytotoxicity in vitro. Moreover, FGF showed a protective effect against cognitive impairment in Aß-treated mice. Molecular dynamics simulations further showed that FGF could synergistically interact with the Aß17-42 pentamer via electrostatic interactions, hydrogen bonds and π-π interactions, which reduced the ß-sheet content, and disordered random coils and bend structures of the Aß17-42 pentamer. This study offers a comprehensive understanding of the inhibitory effects of FGF against Aß neurotoxicity, which is critical for the search of effective food additives that can combat amyloid-associated disease.


Subject(s)
Amyloid beta-Peptides/drug effects , Amyloid/antagonists & inhibitors , Cognitive Dysfunction/prevention & control , Food Additives/therapeutic use , Lissamine Green Dyes/therapeutic use , Neuroprotective Agents/therapeutic use , Protein Aggregation, Pathological/prevention & control , Alzheimer Disease/metabolism , Alzheimer Disease/prevention & control , Amyloid/drug effects , Amyloid/toxicity , Amyloid/ultrastructure , Amyloid beta-Peptides/chemistry , Animals , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Exploratory Behavior/drug effects , Food Additives/pharmacology , Humans , Hydrogen Bonding , Lissamine Green Dyes/pharmacology , Mice , Microscopy, Atomic Force , Models, Molecular , Molecular Dynamics Simulation , Morris Water Maze Test/drug effects , Neuroprotective Agents/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/drug effects , Protein Aggregation, Pathological/drug therapy , Protein Structure, Secondary/drug effects , Random Allocation , Static Electricity
5.
Int Immunopharmacol ; 81: 106263, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32028243

ABSTRACT

Pathological ß-amyloid (Aß)-induced microglial activation could cause chronic neuroinflammation in the brain of Alzheimer's disease (AD) patients, and has been considered as one of the main pathological events of this disease. Chicago sky blue 6B (CSB6B), a pigment used in biochemical staining, has been reported to produce analgesic effects in neuroinflammatory-associated pain models. We have previously found that CSB6B could directly inhibit Aß aggregation and prevent Aß toxicity in neurons. However, it remains unclear whether this compound could prevent Aß-induced neuroinflammation and impairments of learning and memory in the AD models. In this study, CSB6B was found to effectively inhibit the production of pro-inflammatory cytokines, including tumor necrosis factor-α and interleukin-1ß, without affecting cell viability in BV2 microglia cells stimulated by Aß oligomer and lipopolysaccharide. Moreover, CSB6B significantly reduced mRNA expression of inducible nitric oxide synthase and increased mRNA expression of arginase-1, suggesting that CSB6B might promote the polarization of BV2 cells into M2 phenotype. In Aß oligomer-treated mice, hippocampal injection of CSB6B prevented cognitive impairments, and attenuated pro-inflammatory cytokines production. In addition, CSB6B inhibited nuclear transcription factor-κB (NF-κB), and restrainedthe activation of NOD-like receptor pyrin domain containing-3 (NLRP3) both in vitro and in vivo. According to our results, CSB6B may counteract Aß-induced cognitive impairments and neuroinflammation by inhibiting NF-κB and NLRP3. Combined with previous studies, we anticipated that CSB6B may further develop into a potential anti-AD drug with multiple functions on neurons and microglia cells, concurrently.


Subject(s)
Alzheimer Disease/drug therapy , Anti-Inflammatory Agents/therapeutic use , Cognitive Dysfunction/drug therapy , Microglia/metabolism , Neurogenic Inflammation/drug therapy , Neurons/metabolism , Trypan Blue/therapeutic use , Amyloid beta-Peptides/immunology , Animals , Cell Line , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Mice , Mice, Inbred ICR , Microglia/pathology , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neurons/pathology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism
6.
Metab Brain Dis ; 35(2): 373-383, 2020 02.
Article in English | MEDLINE | ID: mdl-31916204

ABSTRACT

Postoperative cognitive dysfunction (POCD) is a common neurological disease affecting the elderly patients after surgery. Unfortunately, no effective treatment for this disease has been discovered. Edaravone, a clinical-used free radical scavenger, at 3 mg/kg has been reported to prevent neuroinflammation induced by the combination of surgery and lipopolysaccharide in adult rodents. However, we found that edaravone at such low concentration could not inhibit POCD in aged mice. Instead, edaravone at 33.2 mg/kg significantly prevented recognition and spatial cognitive dysfunctions in 14 month aged mice after abdominal surgery under general anesthesia with isoflurane. Furthermore, edaravone significantly prevented the increase of tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß) and interleukin-6 (IL-6) induced by abdominal surgery in aged mice. Edaravone could also decrease glial fibrillary acidic protein (GFAP) and ionized calcium binding adaptor molecule-1 (Iba-1) positive areas in the hippocampal regions of surgery mice, suggesting that edaravone might inhibit surgery-induced over-activation of microglia and astrocytes. Moreover, edaravone substantially increased the expression of PSD-95 and pSer9-glycogen synthase kinase-3ß (pSer9-GSK3ß) as demonstrated by Western blotting assay. Furthermore, the activity of acetylcholinesterase (AChE) is decreased in the mice in edaravone group. All these results suggested that edaravone at high concentrations could inhibit surgery-induced cognitive impairments in aged animals, possibly via the attenuation of neuroinflammation, the increase of synaptic proteins, and the elevation of cholinergic transmission, providing a further support that edaravone might be developed as a treatment of POCD.


Subject(s)
Aging/drug effects , Anesthesia, General/adverse effects , Cognitive Dysfunction/prevention & control , Edaravone/administration & dosage , Neuroprotective Agents/administration & dosage , Postoperative Cognitive Complications/prevention & control , Abdominal Wall/surgery , Aging/psychology , Animals , Cognitive Dysfunction/etiology , Cognitive Dysfunction/psychology , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred ICR , Postoperative Cognitive Complications/etiology , Postoperative Cognitive Complications/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...