Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Nutrients ; 15(23)2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38068844

ABSTRACT

Alzheimer's disease (AD), is a progressive neurodegenerative disorder that involves the deposition of ß-amyloid plaques and the clinical symptoms of confusion, memory loss, and cognitive dysfunction. Despite enormous progress in the field, no curative treatment is available. Therefore, the current study was designed to determine the neuroprotective effects of N-methyl-(2S, 4R)-Trans-4-hydroxy-L-proline (NMP) obtained from Sideroxylon obtusifolium, a Brazilian folk medicine with anti-inflammatory and anti-oxidative properties. Here, for the first time, we explored the neuroprotective role of NMP in the Aß1-42-injected mouse model of AD. After acclimatization, a single intracerebroventricular injection of Aß1-42 (5 µL/5 min/mouse) in C57BL/6N mice induced significant amyloidogenesis, reactive gliosis, oxidative stress, neuroinflammation, and synaptic and memory deficits. However, an intraperitoneal injection of NMP at a dose of (50 mg/kg/day) for three consecutive weeks remarkably decreased beta secretase1 (BACE-1) and Aß, activated the astrocyte and microglia expression level as well as downstream inflammatory mediators such as pNF-ĸB, TNF-α, and IL-1ß. NPM also strongly attenuated oxidative stress, as evaluated by the expression level of NRF2/HO-1, and synaptic failure, by improving the level of both the presynaptic (SNAP-25 and SYN) and postsynaptic (PSD-95 and SNAP-23) regions of the synapses in the cortexes and hippocampi of the Aß1-42-injected mice, contributing to cognitive improvement in AD and improving the behavioral deficits displayed in the Morris water maze and Y-maze. Overall, our data suggest that NMP provides potent multifactorial effects, including the inhibition of amyloid plaques, oxidative stress, neuroinflammation, and cognitive deficits.


Subject(s)
Alzheimer Disease , Neuroprotective Agents , Mice , Animals , Alzheimer Disease/chemically induced , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Neuroprotective Agents/therapeutic use , Neuroinflammatory Diseases , Plaque, Amyloid , Mice, Inbred C57BL , Amyloid beta-Peptides/metabolism , Memory Disorders/metabolism , Disease Models, Animal
2.
Nutrients ; 15(13)2023 Jul 07.
Article in English | MEDLINE | ID: mdl-37447385

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disease illustrated by neuronal dysfunctions, leading to memory weaknesses and personality changes mostly in the aged population worldwide. The exact cause of AD is unclear, but numerous studies have addressed the involvement of oxidative stress (OS), induced by reactive oxygen species (ROS), to be one of the leading causes in developing AD. OS dysregulates the cellular homeostasis, causing abnormal protein and lipid metabolism. Nutrition plays a pivotal role in modulating the antioxidant system and decreases the neuronal ROS level, thus playing an important therapeutic role in neurodegenerative diseases, especially in AD. Hence, medicinal herbs and their extracts have received global attention as a commercial source of antioxidants Lupeol. Lupeol is a pentacyclic triterpenoid and has many biological functions. It is available in fruits, vegetables, and medicinal plants. It has shown effective antioxidant and anti-inflammatory properties, and higher blood-brain barrier permeability. Also, the binding and inhibitory potentials of Lupeol have been investigated and proved to be effective against certain receptor proteins and enzymes in AD studies by computational molecular docking approaches. Therefore, AD-related research has gained interest in investigating the therapeutic effects of Lupeol. However, despite its beneficial effects in AD, there is still a lack of research in Lupeol. Hence, we compiled in this analysis all preclinical research that looked at Lupeol as an antioxidant and anti-inflammatory agent for AD.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Triterpenes , Humans , Aged , Antioxidants/pharmacology , Antioxidants/therapeutic use , Antioxidants/metabolism , Alzheimer Disease/metabolism , Reactive Oxygen Species/metabolism , Neurodegenerative Diseases/drug therapy , Triterpenes/pharmacology , Triterpenes/therapeutic use , Molecular Docking Simulation , Oxidative Stress , Pentacyclic Triterpenes/pharmacology , Pentacyclic Triterpenes/therapeutic use
3.
In Vivo ; 36(4): 1753-1760, 2022.
Article in English | MEDLINE | ID: mdl-35738599

ABSTRACT

BACKGROUND/AIM: Dark tea, made by fermentation of tea leaves using microorganisms, is well known for its antiobesity effect; however, studies to identify this effect have not been sufficiently conducted. Herein, the anti-obesity effects of post-fermented dark tea were studied in high-fat diet mouse. MATERIALS AND METHODS: Obesity was induced through a high-fat diet in C57BL/6 mice, and then dark tea extract powder (DTP) was orally administered daily for 12 weeks to evaluate the body and organ weights. Changes in the biochemical markers of obesity were evaluated to study the mechanism of the anti-obesity effects of DTP. RESULTS: When DTP was administered to obesity mice, the weight and food intake reduced, blood aspartate aminotransferase (AST), alanine aminotransferase (ALT), triglyceride (TG), low-density lipoprotein-cholesterol (LDL-C) decreased, whereas high-density lipoprotein cholesterol (HDL-C) increased. Histopathology showed that steatosis and inflammation scores were reduced within the liver and adipocyte sizes were reduced within epididymal adipocyte. In addition, a significant decrease in blood insulin and hepatic TG and a significant increase in blood adiponectin were also confirmed. The results of western blot and qPCR in week 12, showed a significant decrease in the mRNA and protein levels of C/EBPα, and the mRNA levels of PPARγ in the liver. CONCLUSION: Dark tea extracts are thought to have an anti-obesity effect by reducing the levels of the main transcription factors that promote adipocyte differentiation, such as C/EBPα, and PPARγ. Therefore, diet products using dark tea extracts could be developed.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha , PPAR gamma , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-alpha/pharmacology , Cholesterol , Diet, High-Fat/adverse effects , Down-Regulation , Liver/pathology , Mice , Mice, Inbred C57BL , Obesity/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Plant Extracts/chemistry , Plant Extracts/pharmacology , RNA, Messenger/metabolism , Tea/chemistry , Triglycerides/metabolism , Triglycerides/pharmacology
4.
Int J Mol Sci ; 23(3)2022 Feb 04.
Article in English | MEDLINE | ID: mdl-35163692

ABSTRACT

Recently, the world has been witnessing a global pandemic with no effective therapeutics yet, while cancer continues to be a major disease claiming many lives. The natural compound curcumin is bestowed with multiple medicinal applications in addition to demonstrating antiviral and anticancer activities. In order to elucidate the impact of curcumin on COVID-19 and cancer, the current investigation has adapted several computational techniques to unfold its possible inhibitory activity. Accordingly, curcumin and similar compounds and analogues were retrieved and assessed for their binding affinities at the binding pocket of SARS-CoV-2 main protease and DDX3. The best binding pose was escalated to molecular dynamics simulation (MDS) studies to assess the time dependent stability. Our findings have rendered one compound that has demonstrated good molecular dock score complemented by key residue interactions and have shown stable MDS results inferred by root mean square deviation (RMSD), radius of gyration (Rg), binding mode, hydrogen bond interactions, and interaction energy. Essential dynamics results have shown that the systemadapts minimum energy conformation to attain a stable state. The discovered compound (curA) could act as plausible inhibitor against SARS-CoV-2 and DDX3. Furthermore, curA could serve as a chemical scaffold for designing and developing new compounds.


Subject(s)
Curcumin/analogs & derivatives , Curcumin/pharmacology , SARS-CoV-2/drug effects , Antiviral Agents/pharmacology , Computational Biology/methods , Drug Evaluation, Preclinical/methods , Humans , Molecular Docking Simulation/methods , Molecular Dynamics Simulation , Neoplasms/drug therapy , Protease Inhibitors/pharmacology , Protein Binding/drug effects , SARS-CoV-2/pathogenicity , COVID-19 Drug Treatment
5.
Neurosci Lett ; 773: 136495, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35108588

ABSTRACT

Ischemic stroke is the most common type of stroke and is caused by vascular closure. Chlorogenic acid is a polyphenolic compound that is present in various plants. It is used as a traditional oriental medicine because of its anti-oxidant and anti-inflammatory properties. We investigated whether chlorogenic acid mediates neuroprotective effects by regulating pro-inflammatory proteins. Focal cerebral ischemia was induced through middle cerebral artery occlusion (MCAO) surgery in adult rats. Chlorogenic acid (30 mg/kg) or vehicle was injected into the abdominal cavity 2 h after MCAO. Rats were sacrificed 24 h after MCAO surgery and brain tissues were isolated immediately. MCAO caused histopathological changes in the ischemic cerebral cortex, and chlorogenic acid attenuated these changes. Chlorogenic acid reduced MCAO-induced reactive oxygen species generation and oxidative stress increase in the cerebral cortex. Furthermore, cerebral ischemia increased the expression of ionized calcium-binding adapter molecule-1 (Iba-1) and glial fibrillary acidic protein (GFAP), which are microglia and astrocyte activation markers, respectively. However, chlorogenic acid prevented MCAO-induced these increases. MCAO damage also increased the expression of nuclear factor-κB (NF-κB), interleukin-1ß (IL-1ß), and tumor necrosis factor-α (TNF-α). Chlorogenic acid treatment attenuated these increases caused by MCAO. These proteins are representative pro-inflammatory markers. This study confirmed that chlorogenic acid exerts an anti-oxidative effect and elucidated anti-inflammatory effect through regulating NF-κB, IL-1ß, and TNF-α on cerebral ischemia. Thus, we can suggest that chlorogenic acid has neuroprotective effects by reducing oxidative stress and controlling pro-inflammatory proteins against cerebral ischemic damage.


Subject(s)
Brain Ischemia , Neuroprotective Agents , Animals , Anti-Inflammatory Agents/pharmacology , Brain Ischemia/metabolism , Chlorogenic Acid/pharmacology , Chlorogenic Acid/therapeutic use , Disease Models, Animal , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , NF-kappa B/metabolism , Neuroinflammatory Diseases , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
6.
J Inflamm Res ; 14: 6251-6264, 2021.
Article in English | MEDLINE | ID: mdl-34866924

ABSTRACT

This review covers the preclinical and clinical literature supporting the role of melatonin in the management of brain injury-induced oxidative stress, neuroinflammation, and neurodegeneration, and reviews the past and current therapeutic strategies. Traumatic brain injury (TBI) is a neurodegenerative condition, unpredictably and potentially progressing into chronic neurodegeneration, with permanent cognitive, neurologic, and motor dysfunction, having no standard therapies. Due to its complex and multi-faceted nature, the TBI has highly heterogeneous pathophysiology, characterized by the highest mortality and disability worldwide. Mounting evidence suggests that the TBI induces oxidative and nitrosative stress, which is involved in the progression of chronic and acute neurodegenerative diseases. Defenses against such conditions are mostly dependent on the usage of antioxidant compounds, the majority of whom are ingested as nutraceuticals or as dietary supplements. A large amount of literature is available regarding the efficacy of antioxidant compounds to counteract the TBI-associated damage in animal and cellular models of the TBI and several clinical studies. Collectively, the studies have suggested that TBI induces oxidative stress, by suppressing the endogenous antioxidant system, such as nuclear factor erythroid 2-related factor-2 (Nrf-2) increasing the lipid peroxidation and elevation of oxidative damage. Moreover, elevated oxidative stress may induce neuroinflammation by activating the microglial cells, releasing and activating the inflammatory cytokines and inflammatory mediators, and energy dyshomeostasis. Thus, melatonin has shown regulatory effects against the TBI-induced autophagic dysfunction, regulation of mitogen-activated protein kinases, such as ERK, activation of the NLRP-3 inflammasome, and release of the inflammatory cytokines. The collective findings strongly suggest that melatonin may regulate TBI-induced neurodegeneration, although further studies should be conducted to better facilitate future therapeutic windows.

7.
Oxid Med Cell Longev ; 2021: 6635552, 2021.
Article in English | MEDLINE | ID: mdl-33953832

ABSTRACT

The study was aimed at analyzing the protective effects of gintonin in an amyloid beta- (Aß-) induced Alzheimer's disease (AD) mouse model. For the development of the Aß-induced AD mouse model, the amyloid-ß (Aß 1-42) peptide was stereotaxically injected into the brains of mice. Subsequently, gintonin was administered at a dose of 100 mg/kg/day/per oral (p.o) for four weeks daily, and its effects were evaluated by using western blotting, fluorescence analysis of brain sections, biochemical tests, and memory-related behavioral evaluations. To elucidate the effects of gintonin at the mechanistic level, the activation of endogenous antioxidant mechanisms, as well as the activation of astrocytes, microglia, and proinflammatory mediators such as nuclear factor erythroid 2-related factor 2 (NRF-2) and heme oxygenase-1 (HO-1), was evaluated. In addition, microglial cells (BV-2 cells) were used to analyze the effects of gintonin on microglial activation and signaling mechanisms. Collectively, the results suggested that gintonin reduced elevated oxidative stress by improving the expression of NRF-2 and HO-1 and thereby reducing the generation of reactive oxygen species (ROS) and lipid peroxidation (LPO). Moreover, gintonin significantly suppressed activated microglial cells and inflammatory mediators in the brains of Aß-injected mice. Our findings also indicated improved synaptic and memory functions in the brains of Aß-injected mice after treatment with gintonin. These results suggest that gintonin may be effective for relieving AD symptoms by regulating oxidative stress and inflammatory processes in a mouse model of AD. Collectively, the findings of this preclinical study highlight and endorse the potential, multitargeted protective effects of gintonin against AD-associated oxidative damage, neuroinflammation, cognitive impairment, and neurodegeneration.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/prevention & control , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Glycoproteins/therapeutic use , Oxidative Stress/drug effects , Plant Extracts/therapeutic use , Administration, Oral , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Disease Models, Animal , Male , Mice , Plant Extracts/pharmacology
8.
Mol Neurodegener ; 16(1): 23, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33849621

ABSTRACT

BACKGROUND: Recently, we and other researchers reported that brain metabolic disorders are implicated in Alzheimer's disease (AD), a progressive, devastating and incurable neurodegenerative disease. Hence, novel therapeutic approaches are urgently needed to explore potential and novel therapeutic targets/agents for the treatment of AD. The neuronal adiponectin receptor 1 (AdipoR1) is an emerging potential target for intervention in metabolic-associated AD. We aimed to validate this hypothesis and explore in-depth the therapeutic effects of an osmotin-derived adiponectin-mimetic novel nonapeptide (Os-pep) on metabolic-associated AD. METHODS: We used an Os-pep dosage regimen (5 µg/g, i.p., on alternating days for 45 days) for APP/PS1 in amyloid ß oligomer-injected, transgenic adiponectin knockout (Adipo-/-) and AdipoR1 knockdown mice. After behavioral studies, brain tissues were subjected to biochemical and immunohistochemical analyses. In separate cohorts of mice, electrophysiolocal and Golgi staining experiments were performed. To validate the in vivo studies, we used human APP Swedish (swe)/Indiana (ind)-overexpressing neuroblastoma SH-SY5Y cells, which were subjected to knockdown of AdipoR1 and APMK with siRNAs, treated with Os-pep and other conditions as per the mechanistic approach, and we proceeded to perform further biochemical analyses. RESULTS: Our in vitro and in vivo results show that Os-pep has good safety and neuroprotection profiles and crosses the blood-brain barrier. We found reduced levels of neuronal AdipoR1 in human AD brain tissue. Os-pep stimulates AdipoR1 and its downstream target, AMP-activated protein kinase (AMPK) signaling, in AD and Adipo-/- mice. Mechanistically, in all of the in vivo and in vitro studies, Os-pep rescued aberrant neuronal metabolism by reducing neuronal insulin resistance and activated downstream insulin signaling through regulation of AdipoR1/AMPK signaling to consequently improve the memory functions of the AD and Adipo-/- mice, which was associated with improved synaptic function and long-term potentiation via an AdipoR1-dependent mechanism. CONCLUSION: Our findings show that Os-pep activates AdipoR1/AMPK signaling and regulates neuronal insulin resistance and insulin signaling, which subsequently rescues memory deficits in AD and adiponectin-deficient models. Taken together, the results indicate that Os-pep, as an adiponectin-mimetic novel nonapeptide, is a valuable and promising potential therapeutic candidate to treat aberrant brain metabolism associated with AD and other neurodegenerative diseases.


Subject(s)
Alzheimer Disease/drug therapy , Memory Disorders/prevention & control , Neuroprotective Agents/pharmacology , Receptors, Adiponectin/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Adiponectin/deficiency , Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Amyloid beta-Peptides/genetics , Animals , Cell Line, Tumor , Drug Evaluation, Preclinical , Humans , Insulin Resistance , Male , Maze Learning , Memory Disorders/drug therapy , Memory Disorders/etiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Presenilin-1/genetics , RNA Interference , RNA, Small Interfering/genetics , Receptors, Adiponectin/genetics , Signal Transduction
9.
Cells ; 9(6)2020 06 15.
Article in English | MEDLINE | ID: mdl-32549286

ABSTRACT

Neurodegenerative disorders, namely Parkinson's disease (PD), Huntington's disease (HD), Alzheimer's disease (AD), and multiple sclerosis (MS), are increasingly major health concerns due to the increasingly aged population worldwide. These conditions often share the same underlying pathological mechanisms, including elevated oxidative stress, neuroinflammation, and the aggregation of proteins. Several studies have highlighted the potential to diminish the clinical outcomes of these disorders via the administration of herbal compounds, among which gintonin, a derivative of ginseng, has shown promising results. Gintonin is a noncarbohydrate/saponin that has been characterized as a lysophosphatidic acid receptor (LPA Receptor) ligand. Gintonin may cause a significant elevation in calcium levels [Ca2+]i intracellularly, which promotes calcium-mediated cellular effects via the modulation of ion channels and cell surface receptors, regulating the inflammatory effects. Years of research have suggested that gintonin has antioxidant and anti-inflammatory effects against different models of neurodegeneration, and these effects may be employed to tackle the neurological changes. Therefore, we collected the main scientific findings and comprehensively presented them, covering preparation, absorption, and receptor-mediated functions, including effects against Alzheimer's disease models, Parkinson's disease models, anxiety and depression-like models, and other neurological disorders, aiming to provide some insights for the possible usage of gintonin in the management of neurodegenerative conditions.


Subject(s)
Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Plant Extracts/pharmacology , Receptors, Lysophosphatidic Acid/drug effects , Calcium/metabolism , Humans , Ligands , Panax/metabolism , Plant Extracts/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction/drug effects
10.
Cells ; 9(3)2020 03 09.
Article in English | MEDLINE | ID: mdl-32182943

ABSTRACT

In this work, we evaluated the effects of alpha linoleic acid (ALA), an omega-3 polyunsaturated fatty acid, on amyloid-beta-induced glial-cell-mediated neuroinflammation, amyloidogenesis, and cognitive dysfunction in mice. After an infusion of Aß1-42 (Aß1-42, 5 µL/5 min/mouse, intracerebroventricular injection (i.c.v), and respective treatments of ALA (60 mg/kg per oral for six weeks), neuroinflammation, apoptotic markers, and synaptic markers were evaluated by Western blot and immunofluorescence analyses. According to our findings, the infusion of Aß1-42 activated Toll-like receptor 4 (TLR4), glial fibrillary acidic protein (GFAP), and ionized calcium adaptor molecule 1 (Iba-1) in the frontal cortices and hippocampi of the Aß1-42-injected mice to a greater extent than the Aß1-42 + ALA-cotreated mice. Similarly, there was an elevated expression of phospho-c-Jun-N-terminal kinase (p-JNK), phospho-nuclear factor-kB p65 (p-NF-kB p65 (Ser536)), and tissue necrosis factor (TNF) in the Aß1-42 infused mouse brains; interestingly, these markers were significantly reduced in the Aß + ALA-cotreated group. The elevated expression of pro-apoptotic markers was observed during apoptotic cell death in the Aß1-42-treated mouse brains, whereas these markers were markedly reduced in the Aß + ALA-cotreated group. Moreover, Aß1-42 infusion significantly increased amyloidogenesis, as assessed by the enhanced expression of the amyloid precursor proteins (APP) beta-amyloid cleaving enzyme-1 (BACE-1) and amyloid-beta (Aß1-42) in the mouse brains, whereas these proteins were markedly reduced in the Aß + ALA-cotreated group. We also checked the effects of ALA against Aß-triggered synaptic dysfunction and memory dysfunction, showing that ALA significantly improved memory and synaptic functions in Aß-treated mouse brains. These results indicated that ALA could be an applicable intervention in neuroinflammation, apoptotic cell loss, amyloidogenesis, and memory dysfunction via the inhibition of TLR4 and its downstream targets in Aß + ALA-cotreated mouse brains.


Subject(s)
Cognitive Dysfunction/drug therapy , Inflammation/drug therapy , Linoleic Acid/pharmacology , Microglia/drug effects , Administration, Oral , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Animals , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism , Disease Models, Animal , Linoleic Acid/administration & dosage , Linoleic Acid/metabolism , Male , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology
11.
Oxid Med Cell Longev ; 2019: 7860650, 2019.
Article in English | MEDLINE | ID: mdl-31827700

ABSTRACT

Curcumin is a natural polyphenolic compound widely known to have antioxidant, anti-inflammatory, and antiapoptotic properties. In the present study, we explored the neuroprotective effect of curcumin against lipopolysaccharide- (LPS-) induced reactive oxygen species- (ROS-) mediated neuroinflammation, neurodegeneration, and memory deficits in the adult rat hippocampus via regulation of the JNK/NF-κB/Akt signaling pathway. Adult rats were treated intraperitoneally with LPS at a dose of 250 µg/kg for 7 days and curcumin at a dose of 300 mg/kg for 14 days. After 14 days, the rats were sacrificed, and western blotting and ROS and lipid peroxidation assays were performed. For immunohistochemistry and confocal microscopy, the rats were perfused transcardially with 4% paraformaldehyde. In order to verify the JNK-dependent neuroprotective effect of curcumin and to confirm the in vivo results, HT-22 neuronal and BV2 microglial cells were exposed to LPS at a dose of 1 µg/ml, curcumin 100 µg/ml, and SP600125 (a specific JNK inhibitor) 20 µM. Our immunohistochemical, immunofluorescence, and biochemical results revealed that curcumin inhibited LPS-induced oxidative stress by reducing malondialdehyde and 2,7-dichlorofluorescein levels and ameliorating neuroinflammation and neuronal cell death via regulation of the JNK/NF-κB/Akt signaling pathway both in vivo (adult rat hippocampus) and in vitro (HT-22/BV2 cell lines). Moreover, curcumin markedly improved LPS-induced memory impairment in the Morris water maze and Y-maze tasks. Taken together, our results suggest that curcumin may be a potential preventive and therapeutic candidate for LPS-induced ROS-mediated neurotoxicity and memory deficits in an adult rat model.


Subject(s)
Curcumin/pharmacology , Inflammation/prevention & control , Lipopolysaccharides/toxicity , Memory Disorders/prevention & control , Neurotoxicity Syndromes/prevention & control , Signal Transduction/drug effects , Synaptic Transmission/drug effects , Animals , Antioxidants/pharmacology , Dietary Supplements , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Memory Disorders/chemically induced , Memory Disorders/metabolism , Memory Disorders/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley
12.
J Clin Med ; 8(6)2019 Jun 14.
Article in English | MEDLINE | ID: mdl-31207963

ABSTRACT

Phytomedicine based natural flavonoids have potent antioxidant, anti-inflammatory, and neuroprotective activities against neurodegenerative diseases. The aim of the present study is to investigate the potent neuroprotective and antioxidant potential effects of fisetin (natural flavonoid) against central nervous system (CNS)-insult, lipopolysaccharide (LPS)-induced reactive oxygen species (ROS), neuroinflammation, neurodegeneration, and synaptic/memory deficits in adult mice. The mice were injected intraperitoneally (i.p.) with LPS (250 µg/kg/day for 1 week) and a fisetin dosage regimen (20 mg/kg/day i.p. for 2 weeks, 1 week pre-treated to LPS and 1 week co-treated with LPS). Behavioral tests, and biochemical and immunofluorescence assays were applied. Our results revealed that fisetin markedly abrogated the LPS-induced elevated ROS/oxidative stress and activated phosphorylated c-JUN N-terminal Kinase (p-JNK) in the adult mouse hippocampus. Fisetin significantly alleviated LPS-induced activated gliosis. Moreover, fisetin treatment inhibited LPS-induced activation of the inflammatory Toll-like Receptors (TLR4)/cluster of differentiation 14 (CD14)/phospho-nuclear factor kappa (NF-κB) signaling and attenuated other inflammatory mediators (tumor necrosis factor-α (TNF-α), interleukin-1 ß (IL1-ß), and cyclooxygenase (COX-2). Furthermore, immunoblotting and immunohistochemical results revealed that fisetin significantly reversed LPS-induced apoptotic neurodegeneration. Fisetin improved the hippocampal-dependent synaptic and memory functions in LPS-treated adult mice. In summary, our results strongly recommend that fisetin, a natural potent antioxidant, and neuroprotective phytomedicine, represents a promising, valuable, and therapeutic candidate for the prevention and treatment of neurodegenerative diseases.

13.
Nutrients ; 11(6)2019 May 28.
Article in English | MEDLINE | ID: mdl-31141884

ABSTRACT

All over the world, metabolic syndrome constitutes severe health problems. Multiple factors have been reported in the pathogenesis of metabolic syndrome. Metabolic disorders result in reactive oxygen species (ROS) induced oxidative stress, playing a vital role in the development and pathogenesis of major health issues, including neurological disorders Alzheimer's disease (AD) Parkinson's disease (PD). Considerable increasing evidence indicates the substantial contribution of ROS-induced oxidative stress in neurodegenerative diseases. An imbalanced metabolism results in a defective antioxidant defense system, free radicals causing inflammation, cellular apoptosis, and tissue damage. Due to the annual increase in financial and social burdens, in addition to the adverse effects associated with available synthetic agents, treatment diversion from synthetic to natural approaches has occurred. Antioxidants are now being considered as convincing therapeutic agents against various neurodegenerative disorders. Therefore, medicinal herbs and fruits currently receive substantially more attention as commercial sources of antioxidants. In this review, we argue that ROS-targeted therapeutic interventions with naturally occurring antioxidant flavonoid, anthocyanin, and anthocyanin-loaded nanoparticles might be the ultimate treatment against devastating illnesses. Furthermore, we elucidate the hidden potential of the neuroprotective role of anthocyanins and anthocyanin-loaded nanoparticles in AD and PD neuropathies, which lack sufficient attention compared with other polyphenols, despite their strong antioxidant potential. Moreover, we address the need for future research studies of native anthocyanins and nano-based-anthocyanins, which will be helpful in developing anthocyanin treatments as therapeutic mitochondrial antioxidant drug-like regimens to delay or prevent the progression of neurodegenerative diseases, such as AD and PD.


Subject(s)
Anthocyanins/therapeutic use , Antioxidants/therapeutic use , Brain/drug effects , Metabolic Syndrome/drug therapy , Nerve Degeneration , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Animals , Anthocyanins/adverse effects , Antioxidants/adverse effects , Brain/metabolism , Brain/pathology , Humans , Inflammation Mediators/metabolism , Metabolic Syndrome/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neuroprotective Agents/adverse effects , Reactive Oxygen Species/metabolism , Signal Transduction
14.
Nutrients ; 11(5)2019 May 15.
Article in English | MEDLINE | ID: mdl-31096703

ABSTRACT

The aim of the current study was to explore the underlying neuroprotective mechanisms of curcumin (50 mg/kg, for six weeks) against ethanol (5 mg/kg i.p., for six weeks) induced oxidative stress and inflammation-mediated cognitive dysfunction in mice. According to our findings, ethanol triggered reactive oxygen species (ROS), apoptosis, neuroinflammation, and memory impairment, which were significantly inhibited with the administration of curcumin, as assessed by ROS, lipid peroxidation (LPO), and Nrf2/HO-1 (nuclear factor erythroid 2-related factor 2/Heme-oxygenase-1) expression in the experimental mice brains. Moreover, curcumin regulated the expression of the glial cell markers in ethanol-treated mice brains, as analyzed by the relative expression TLR4 (Toll like Receptor 4), RAGE (Receptor for Advanced Glycations End products), GFAP (Glial fibrillary acidic protein), and Iba-1 (Ionized calcium binding adaptor molecule 1), through Western blot and confocal microscopic analysis. Moreover, our results showed that curcumin downregulated the expression of p-JNK (Phospo c-Jun N-Terminal Kinase), p-NF-kB (nuclear factor kappa-light-chain-enhancer of activated B cells), and its downstream targets, as assessed by Western blot and confocal microscopic analysis. Finally, the expression of synaptic proteins and the behavioral results also supported the hypothesis that curcumin may inhibit memory dysfunction and behavioral alterations associated with ethanol intoxication. Altogether, to the best of our knowledge, we believe that curcumin may serve as a potential, promising, and cheaply available neuroprotective compound against ethanol-associated neurodegenerative diseases.


Subject(s)
Curcumin/pharmacology , Dietary Supplements , Memory Disorders/chemically induced , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Animals , Cell Line , Gene Expression Regulation/drug effects , Hippocampus/cytology , Lipid Peroxidation , Male , Memory Disorders/prevention & control , Mice , Mice, Inbred C57BL , Microglia/drug effects , NF-E2-Related Factor 2/genetics , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/prevention & control , Neurons/drug effects , RNA Interference , RNA, Small Interfering , Reactive Oxygen Species , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
15.
Mol Neurobiol ; 56(1): 39-55, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29675576

ABSTRACT

Gintonin, a ginseng-derived glycolipoprotein isolated from ginseng, has been shown to be neuroprotective in several neurological disorders such as Alzheimer's disease models and depressive-like behaviors. In this study, we sought to investigate the potential protective mechanisms of gintonin in an in vivo MPTP and in vitro MPP+-mediated Parkinson's disease (PD) model. We hypothesized that activation of nuclear factor erythroid 2-related factor 2/heme oxygenase-1 (Nrf2/HO-1, potential therapeutic targets for neurodegeneration) with gintonin could abrogate PD-associated neurotoxicity by modulating the accumulation of α-synuclein, neuroinflammation, and apoptotic cell death in an MPTP/MPP+ models of PD. Our in vivo and in vitro findings suggest that the neuroprotective effects of gintonin were associated with the regulation of the Nrf2/HO-1 pathway, which regulated the expression of proinflammatory cytokines and nitric oxide synthase and apoptotic markers in the substantia nigra and striatum of the mice. Moreover, the neuroprotective effects of gintonin were also associated with a reduction in α-synuclein accumulation in the mouse substantia nigra and striatum. The neuroprotective effects of gintonin were further validated by analyzing the effects of gintonin on MPP+-treated SH-SY5Y cells, which confirmed the protective effects of gintonin. It remains for future basic and clinical research to determine the potential use of gintonin in Parkinson's disease. However, to the best of our knowledge, marked alterations in biochemical and morphological setup of midbrain dopaminergic pathways by gintonin in MPTP mice model have not been previously reported. We believe that gintonin might be explored as an important therapeutic agent in the treatment of PD.


Subject(s)
Corpus Striatum/pathology , Dopaminergic Neurons/pathology , Heme Oxygenase-1/metabolism , NF-E2-Related Factor 2/metabolism , Plant Extracts/pharmacology , Signal Transduction , Substantia Nigra/pathology , alpha-Synuclein/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Cell Line, Tumor , Corpus Striatum/physiopathology , Disease Models, Animal , Dopaminergic Neurons/metabolism , Glial Fibrillary Acidic Protein/metabolism , Gliosis/complications , Gliosis/pathology , Gliosis/physiopathology , Humans , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Motor Activity/drug effects , Neuroprotective Agents/pharmacology , Neurotoxins/toxicity , Oxidative Stress/drug effects , Parkinson Disease/complications , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Rotenone , Signal Transduction/drug effects , Substantia Nigra/physiopathology , Tyrosine 3-Monooxygenase/metabolism , Up-Regulation/drug effects
16.
Mol Neurobiol ; 55(7): 6076-6093, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29170981

ABSTRACT

Well-established studies have shown an elevated level of reactive oxygen species (ROS) that induces oxidative stress in the Alzheimer's disease (AD) patient's brain and an animal model of AD. Herein, we investigated the underlying anti-oxidant neuroprotective mechanism of natural dietary supplementation of anthocyanins extracted from Korean black beans in the amyloid precursor protein/presenilin-1 (APP/PS1) mouse model of AD. Both in vivo (APP/PS1 mice) and in vitro (mouse hippocampal HT22 cells) results demonstrated that anthocyanins regulate the phosphorylated-phosphatidylinositol 3-kinase-Akt-glycogen synthase kinase 3 beta (p-PI3K/Akt/GSK3ß) pathways and consequently attenuate amyloid beta oligomer (AßO)-induced elevations in ROS level and oxidative stress via stimulating the master endogenous anti-oxidant system of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (Nrf2/HO-1) pathways and prevent apoptosis and neurodegeneration by suppressing the apoptotic and neurodegenerative markers such as activation of caspase-3 and PARP-1 expression as well as the TUNEL and Fluoro-Jade B-positive neuronal cells in the APP/PS1 mice. In vitro ApoTox-Glo™ Triplex assay results also showed that anthocyanins act as a potent anti-oxidant neuroprotective agent and reduce AßO-induced neurotoxicity in the HT22 cells via PI3K/Akt/Nrf2 signaling. Importantly, anthocyanins improve memory-related pre- and postsynaptic protein markers and memory functions in the APP/PS1 mice. In conclusion, our data suggested that consumption and supplementation of natural-derived anti-oxidant neuroprotective agent such as anthocyanins may be beneficial and suggest new dietary-supplement strategies for intervention in and prevention of progressive neurodegenerative diseases, such as AD.


Subject(s)
Alzheimer Disease/drug therapy , Anthocyanins/therapeutic use , Dietary Supplements , Memory Disorders/drug therapy , Nerve Degeneration/drug therapy , Oxidative Stress , Signal Transduction , Alzheimer Disease/complications , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Anthocyanins/pharmacology , Biomarkers/metabolism , Cell Line , Cell Nucleus/metabolism , Disease Models, Animal , Gene Expression Regulation , Heme Oxygenase-1/metabolism , Humans , Male , Memory Disorders/complications , Memory Disorders/genetics , Memory Disorders/physiopathology , Mice, Inbred C57BL , Mice, Transgenic , NF-E2-Related Factor 2/metabolism , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism
17.
Curr Pharm Biotechnol ; 18(10): 849-854, 2017.
Article in English | MEDLINE | ID: mdl-29205116

ABSTRACT

BACKGROUND: Rhododendron brachycarpum (RB) is a genus of flowering plants generally used for traditional medicine in Korea to treat hypertension, neuralgia, and sterility. Previous studies have shown that RB extract alleviate inflammation and antimicrobial activity. OBJECTIVE: In this study, the effects of RB and its different fractions (n-hexane, ethyl-acetate and n-butanol) on antioxidant activity, DNA damage prevention and the activity of α-glucosidase were studied. RESULTS: The antioxidant ability of RB was investigated in vitro, including that of DPPH-radical and reducing power. As expected, scavenging effect against DPPH-radical of ethyl acetate fraction (IC50 = 17.7 ± 0.5 µg/ml) of RB had the highest DPPH radical scavenging activity, and it was superior to the positive control, butylated hydroxytoluene (BHT) (IC50 = 80.8 ± 1.5 µg/ml). And the reducing power of RB was 3.18 at 1.0 mg/ml. Meanwhile, the α-glucosidase inhibitory activity and prevention of oxidation stress-induced DNA damage were also highest in the ethyl acetate fraction. Pretreatment of pancreatic ß-cells from Syrian golden hamster (HIT-TI5) with the ethyl acetate fraction at concentrations of 300 µg/ml significantly protected the cells from high glucose-induced cell death. CONCLUSION: Our results indicate that ethyl acetate fraction of RB leaves extract has strong antioxidant, α-glucosidase, and prevention of DNA damage activities, and furthermore, ethyl acetate fraction significantly protected the cells from high glucose-induced cell death.


Subject(s)
Antioxidants/pharmacology , Glycoside Hydrolase Inhibitors/pharmacology , Insulin-Secreting Cells/drug effects , Plant Extracts/pharmacology , Rhododendron/chemistry , alpha-Glucosidases/metabolism , Animals , Antioxidants/isolation & purification , Biphenyl Compounds/chemistry , Cell Line , Cell Survival/drug effects , DNA Damage , Glycoside Hydrolase Inhibitors/isolation & purification , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/metabolism , Mesocricetus , Oxidative Stress/drug effects , Oxidative Stress/genetics , Picrates/chemistry , Plant Extracts/isolation & purification , Plant Leaves/chemistry
18.
J Neuroinflammation ; 13(1): 286, 2016 11 08.
Article in English | MEDLINE | ID: mdl-27821173

ABSTRACT

BACKGROUND: Glutamate-induced excitotoxicity, oxidative damage, and neuroinflammation are believed to play an important role in the development of a number of CNS disorders. We recently reported that a high dose of glutamate could induce AMPK-mediated neurodegeneration in the postnatal day 7 (PND7) rat brain. Yet, the mechanism of glutamate-induced oxidative stress and neuroinflammation in the postnatal brain is not well understood. Here, we report for the first time the mechanism of glutamate-induced oxidative damage, neuroinflammation, and neuroprotection by polyphenolic anthocyanins in PND7. METHODS: PND7 rat brains, SH-SY5Y, and BV2 cells treated either alone with glutamate or in combination with anthocyanins and compound C were examined with Western blot and immunofluorescence techniques. Additionally, reactive oxygen species (ROS) assay and other ELISA kit assays were employed to know the therapeutic efficacy of anthocyanins against glutamate. RESULTS: A single injection of glutamate to developing rats significantly increased brain glutamate levels, activated and phosphorylated AMPK induction, and inhibited nuclear factor-E2-related factor 2 (Nrf2) after 2, 3, and 4 h in a time-dependent manner. In contrast, anthocyanin co-treatment significantly reduced glutamate-induced AMPK induction, ROS production, neuroinflammation, and neurodegeneration in the developing rat brain. Most importantly, anthocyanins increased glutathione (GSH and GSSG) levels and stimulated the endogenous antioxidant system, including Nrf2 and heme oxygenase-1 (HO-1), against glutamate-induced oxidative stress. Interestingly, blocking AMPK with compound C in young rats abolished glutamate-induced neurotoxicity. Similarly, all these experiments were replicated in SH-SY5Y cells by silencing AMPK with siRNA, which suggests that AMPK is the key mediator in glutamate-induced neurotoxicity. CONCLUSIONS: Here, we report for the first time that anthocyanins can potentially decrease glutamate-induced neurotoxicity in young rats. Our work demonstrates that glutamate is toxic to the developing rat brain and that anthocyanins can minimize the severity of glutamate-induced neurotoxicity in an AMPK-dependent manner.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Anthocyanins/pharmacology , Anthocyanins/therapeutic use , Encephalitis/drug therapy , Nerve Degeneration/drug therapy , Oxidative Stress/drug effects , AMP-Activated Protein Kinases/genetics , Animals , Animals, Newborn , Antioxidants/pharmacology , Antioxidants/therapeutic use , Cell Line, Transformed , Cyclooxygenase 2/metabolism , Encephalitis/chemically induced , Enzyme Activation/drug effects , Gene Expression Regulation, Developmental/drug effects , Glutamine/toxicity , Humans , Nerve Degeneration/chemically induced , Neuroblastoma/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
19.
J Surg Res ; 201(1): 141-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26850195

ABSTRACT

BACKGROUND: Curcumin is known to have a neuroprotective effect against cerebral ischemia. The objective of this study was to identify various proteins that are differentially expressed by curcumin treatment in focal cerebral ischemia using a proteomic approach. METHODS: Adult male rats were treated with vehicle or curcumin 1 h after middle cerebral artery occlusion. Brain tissues were collected 24 h after the onset of middle cerebral artery occlusion, and cerebral cortices proteins were identified by two-dimensional gel electrophoresis and mass spectrometry. RESULTS: We detected several proteins with altered expression levels between vehicle- and curcumin-treated animals. Among these proteins, ubiquitin carboxy-terminal hydrolase L1, isocitrate dehydrogenase, adenosylhomocysteinase, and eukaryotic initiation factor 4A were decreased in the vehicle-treated animal, and curcumin treatment attenuated the injury-induced decreases of these proteins. Conversely, pyridoxal phosphate phosphatase was increased in the vehicle-treated animal, and curcumin treatment prevented decreases in this protein. The identified altered proteins are associated with cellular metabolism and differentiation. CONCLUSIONS: The results of this study suggest that curcumin exerts a neuroprotective effect by regulating the expression of various proteins in focal cerebral ischemia.


Subject(s)
Brain/drug effects , Curcumin/therapeutic use , Gene Expression Regulation/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/therapeutic use , Animals , Brain/metabolism , Curcumin/pharmacology , Electrophoresis, Gel, Two-Dimensional , Infarction, Middle Cerebral Artery/metabolism , Male , Mass Spectrometry , Neuroprotective Agents/pharmacology , Phytotherapy , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Random Allocation , Rats, Sprague-Dawley
20.
Nanoscale ; 7(37): 15225-37, 2015 Oct 07.
Article in English | MEDLINE | ID: mdl-26315713

ABSTRACT

The adverse effects of nanoscale-alumina (Al2O3-NPs) have been previously demonstrated in both in vitro and in vivo studies, whereas little is known about their mechanism of neurotoxicity. It is the goal of this research to determine the toxic effects of nano-alumina on human neuroblastoma SH-SY5Y and mouse hippocampal HT22 cells in vitro and on ICR female mice in vivo. Nano-alumina displayed toxic effects on SH-SY5Y cell lines in three different concentrations also increased aluminium abundance and induced oxidative stress in HT22 cells. Nano-alumina peripherally administered to ICR female mice for three weeks increased brain aluminium and ROS production, disturbing brain energy homeostasis, and led to the impairment of hippocampus-dependent memory. Most importantly, these nano-particles induced Alzheimer disease (AD) neuropathology by enhancing the amyloidogenic pathway of Amyloid Beta (Aß) production, aggregation and implied the progression of neurodegeneration in the cortex and hippocampus of these mice. In conclusion, these data demonstrate that nano-alumina is toxic to both cells and female mice and that prolonged exposure may heighten the chances of developing a neurodegenerative disease, such as AD.


Subject(s)
Aluminum Oxide/toxicity , Amyloid beta-Peptides/metabolism , Metal Nanoparticles/toxicity , Oxidative Stress/drug effects , Aluminum Oxide/chemistry , Animals , Cell Line, Tumor , Cell Survival/drug effects , Female , Humans , Metal Nanoparticles/chemistry , Mice , Mice, Inbred ICR
SELECTION OF CITATIONS
SEARCH DETAIL