Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mol Nutr Food Res ; 65(15): e2000986, 2021 08.
Article in English | MEDLINE | ID: mdl-33974360

ABSTRACT

SCOPE: Studies based on DHA/EPA supplementation in animal models of inflammatory bowel disease (IBD) reveal controversial results. It is speculated that different forms of DHA may explain the controversial results. Therefore, the effects of DHA-enriched phospholipids (DHA-PL) and DHA-enriched triglyceride (DHA-TG) on IBD are compared. METHODS AND RESULTS: Male C57BL6/J mice are given DHA-PL and DHA-TG for 14 consecutive days, and receive ad libitum a 3.0% dextran sodium sulfate solution on the eighth day to establish IBD model. The results show that both DHA-PL and DHA-TG can reverse the colitis pathological process by decreasing the disease activity indexes (DAI), raising the colon length, suppressing the intestinal permeability, suppressing the oxidative stress, down-regulating pro-inflammatory factors, up-regulating anti-inflammatory factor in colon tissues. DHA-PL and DHA-TG also regulate the composition of gut microbiota via decreasing of the abundance Bacteroidetes and Firmicutes, and DHA-TG increases the abundance of Odoribacter. Importantly, DHA-PL and DHA-TG obviously attenuate the activation of microglia. CONCLUSIONS: DHA-PL shows outstanding advantages in regulating oxidative stress, inflammatory responses, and intestinal barrier permeability. The current research indicates that the existence of DHA affects the improvement, DHA in phospholipid form could be a more effective choice for nutritional intervention to prevent and treat colitis.


Subject(s)
Colitis/diet therapy , Encephalitis/diet therapy , Gastrointestinal Microbiome/drug effects , Oxidative Stress/drug effects , Phospholipids/pharmacology , Administration, Oral , Animals , Colitis/chemically induced , Colitis/microbiology , Colitis/pathology , Cytokines/metabolism , Dextran Sulfate/toxicity , Dietary Supplements , Docosahexaenoic Acids/pharmacology , Encephalitis/etiology , Fatty Acids/analysis , Fatty Acids/chemistry , Fatty Acids, Omega-3/chemistry , Fatty Acids, Omega-3/pharmacology , Gastrointestinal Microbiome/genetics , Male , Mice, Inbred C57BL , Phospholipids/administration & dosage
2.
Nutr Neurosci ; 23(8): 640-645, 2020 Aug.
Article in English | MEDLINE | ID: mdl-30404563

ABSTRACT

Backgrounds and aims: Clinical studies demonstrated that the efficacy of Coenzyme Q10 (CoQ10) as an adjuvant therapeutic agent in several neurological diseases such as Parkinson disease (PD), Huntington disease (HD), and migraine. The purpose of this study is to investigate oxidative stress effects, antioxidant enzymes activity, neuroinflammatory markers levels, and neurological outcome in acute ischemic stroke (AIS) patients following administration of CoQ10 (300 mg/day). Methods: Patients with AIS (n = 60) were randomly assigned to a placebo group (wheat starch, n = 30) or CoQ10-supplemented group (300 mg/day, n = 30). The intervention was administered for 4 weeks. Serum CoQ10 concentration, malondialdehyde (MDA), superoxide dismutase (SOD) activity, glial fibrillary acidic protein (GFAP) levels as primary outcomes and National Institute of Health Stroke Scale (NIHSS), Modified Ranking Scale (MRS), and Mini-Mental State Examination (MMSE) as secondary outcome were measured at the both beginning and end of the study. Results: Forty-four subjects with AIS completed the intervention study. A significant increase in CoQ10 level was observed in the supplement-treated group compared with placebo group (mean difference = 26.05 ± 26.63 ng/ml, 14.12 ± 14.69 ng/ml, respectively; P = 0.01), moreover CoQ10 supplementation improved NIHSS and MMSE scores significantly (P = 0.05, P = 0.03 respectively). but there were no statistically significant differences in MRS score, MDA, SOD, and GFAP levels between the two groups. Conclusions: CoQ10 probably due to low dose and short duration of supplementation, no favorable effects on MDA level, SOD activity and GFAP level.


Subject(s)
Brain Ischemia/diet therapy , Neuroprotective Agents/administration & dosage , Stroke/diet therapy , Ubiquinone/analogs & derivatives , Vitamins/administration & dosage , Aged , Antioxidants/metabolism , Brain Ischemia/complications , Brain Ischemia/metabolism , Double-Blind Method , Encephalitis/complications , Encephalitis/diet therapy , Encephalitis/metabolism , Female , Humans , Male , Middle Aged , Oxidative Stress/drug effects , Stroke/complications , Stroke/metabolism , Ubiquinone/administration & dosage
3.
Pharmacol Biochem Behav ; 184: 172743, 2019 09.
Article in English | MEDLINE | ID: mdl-31356838

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is the most common neurodegenerative disease causing dementia in the elderly population. Due to the fact that there is still no cure for Alzheimer's dementia and available treatment strategies bring only symptomatic benefits, there is a pressing demand for other effective strategies such as diet. Since the inflammation hypothesis gained considerable significance in the AD pathogenesis, elucidating the modulatory role of dietary factors on inflammation may help to prevent, delay the onset and slow the progression of AD. Current evidence clearly shows that synergistic action of combined supplementation and complex dietary patterns provides stronger benefits than any single component considered separately. Recent studies reveal the growing importance of novel factors such as dietary advanced glycation end products (d-AGE), gut microbiota, butyrate and vitamin D3 on inflammatory processes in AD. CONCLUSION: This paper summarizes the available evidence of pro- and anti-inflammatory activity of some dietary components including fatty acids, vitamins, flavonoids, polyphenols, probiotics and d-AGE, and their potential for AD prevention and treatment.


Subject(s)
Alzheimer Disease/diet therapy , Alzheimer Disease/prevention & control , Diet , Dietary Supplements , Encephalitis/diet therapy , Butyrates/therapeutic use , Caffeine/therapeutic use , Cholecalciferol/therapeutic use , Curcumin/therapeutic use , Fatty Acids/adverse effects , Fatty Acids, Omega-3/therapeutic use , Gastrointestinal Microbiome/physiology , Glycation End Products, Advanced/adverse effects , Humans , Meat/adverse effects , Resveratrol/therapeutic use , Vitamin B Complex/therapeutic use
4.
Front Immunol ; 9: 1832, 2018.
Article in English | MEDLINE | ID: mdl-30154787

ABSTRACT

Aging results in chronic systemic inflammation that can alter neuroinflammation of the brain. Specifically, microglia shift to a pro-inflammatory phenotype predisposing them to hyperactivation upon stimulation by peripheral immune signals. It is proposed that certain nutrients can delay brain aging by preventing or reversing microglial hyperactivation. Butyrate, a short-chain fatty acid (SCFA) produced primarily by bacterial fermentation of fiber in the colon, has been extensively studied pharmacologically as a histone deacetylase inhibitor and serves as an attractive therapeutic candidate, as butyrate has also been shown to be anti-inflammatory and improve memory in animal models. In this study, we demonstrate that butyrate can attenuate pro-inflammatory cytokine expression in microglia in aged mice. It is still not fully understood, however, if an increase in butyrate-producing bacteria in the gut as a consequence of a diet high in soluble fiber could affect microglial activation during aging. Adult and aged mice were fed either a 1% cellulose (low fiber) or 5% inulin (high fiber) diet for 4 weeks. Findings indicate that mice fed inulin had an altered gut microbiome and increased butyrate, acetate, and total SCFA production. In addition, histological scoring of the distal colon demonstrated that aged animals on the low fiber diet had increased inflammatory infiltrate that was significantly reduced in animals consuming the high fiber diet. Furthermore, gene expression of inflammatory markers, epigenetic regulators, and the microglial sensory apparatus (i.e., the sensome) were altered by both diet and age, with aged animals exhibiting a more anti-inflammatory microglial profile on the high fiber diet. Taken together, high fiber supplementation in aging is a non-invasive strategy to increase butyrate levels, and these data suggest that an increase in butyrate through added soluble fiber such as inulin could counterbalance the age-related microbiota dysbiosis, potentially leading to neurological benefits.


Subject(s)
Aging/immunology , Aging/metabolism , Butyrates/administration & dosage , Dietary Fiber/administration & dosage , Encephalitis/etiology , Aging/genetics , Animals , DNA Methylation , Disease Models, Animal , Encephalitis/diet therapy , Encephalitis/metabolism , Encephalitis/pathology , Epigenesis, Genetic , Gastrointestinal Microbiome , Gene Expression Regulation/drug effects , Hippocampus/immunology , Hippocampus/metabolism , Interleukin-1beta/genetics , Lipopolysaccharides/immunology , Mice , Microglia/immunology , Microglia/metabolism , Tight Junctions/metabolism
5.
J Neuroimmunol ; 310: 111-119, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28778434

ABSTRACT

In patients with Alzheimer's disease (AD), a persistent and unresolved neuroinflammatory process can contribute to neuronal loss and a decline in their cognitive and functional abilities. Recent studies have demonstrated that the ability to resolve inflammation is impaired in the brains of patients with AD. Preclinical evidence demonstrates the potential of therapeutic interventions on the resolution phase of inflammation in AD. Supplementation of omega-3 fatty acids (n-3 FAs), precursors for specialized pro-resolving mediators, emerged as a possibility for prevention and management of AD. Here, we provide a narrative review of resolving inflammation in AD and the role of n-3 FA supplementation in AD.


Subject(s)
Alzheimer Disease/complications , Dietary Supplements , Encephalitis/diet therapy , Encephalitis/etiology , Fatty Acids, Omega-3/metabolism , Alzheimer Disease/prevention & control , Animals , Humans
6.
J Neuroinflammation ; 14(1): 143, 2017 07 24.
Article in English | MEDLINE | ID: mdl-28738820

ABSTRACT

BACKGROUND: Microglial activation and the subsequent inflammatory response in the central nervous system play important roles in secondary damage after traumatic brain injury (TBI). High-mobility group box 1 (HMGB1) protein, an important mediator in late inflammatory responses, interacts with transmembrane receptor for advanced glycation end products (RAGE) and toll-like receptors (TLRs) to activate downstream signaling pathways, such as the nuclear factor (NF)-κB signaling pathway, leading to a cascade amplification of inflammatory responses, which are related to neuronal damage after TBI. Omega-3 polyunsaturated fatty acid (ω-3 PUFA) is a commonly used clinical immunonutrient, which has antioxidative and anti-inflammatory effects. However, the effects of ω-3 PUFA on HMGB1 expression and HMGB1-mediated activation of the TLR4/NF-κB signaling pathway are not clear. METHODS: The Feeney DM TBI model was adopted to induce brain injury in rats. Modified neurological severity scores, brain water content, and Nissl staining were employed to determine the neuroprotective effects of ω-3 PUFA supplementation. Assessment of microglial activation in lesioned sites and protein markers for proinflammatory, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6, interferon (IFN)-γ, and HMGB1 were used to evaluate neuroinflammatory responses and anti-inflammation effects of ω-3 PUFA supplementation. Immunofluorescent staining and western blot analysis were used to detect HMGB1 nuclear translocation, secretion, and HMGB1-mediated activation of the TLR4/NF-κB signaling pathway to evaluate the effects of ω-3 PUFA supplementation and gain further insight into the mechanisms underlying the development of the neuroinflammatory response after TBI. RESULTS: It was found that ω-3 PUFA supplementation inhibited TBI-induced microglial activation and expression of inflammatory factors (TNF-α, IL-1ß, IL-6, and IFN-γ), reduced brain edema, decreased neuronal apoptosis, and improved neurological functions after TBI. We further demonstrated that ω-3 PUFA supplementation inhibited HMGB1 nuclear translocation and secretion and decreased expression of HMGB1 in neurons and microglia in the lesioned areas. Moreover, ω-3 PUFA supplementation inhibited microglial activation and the subsequent inflammatory response by regulating HMGB1 and the TLR4/NF-κB signaling pathway. CONCLUSIONS: The results of this study suggest that microglial activation and the subsequent neuroinflammatory response as well as the related HMGB1/TLR4/NF-κB signaling pathway play essential roles in secondary injury after TBI. Furthermore, ω-3 PUFA supplementation inhibited TBI-induced microglial activation and the subsequent inflammatory response by regulating HMGB1 nuclear translocation and secretion and also HMGB1-mediated activation of the TLR4/NF-κB signaling pathway, leading to neuroprotective effects.


Subject(s)
Brain Injuries, Traumatic/pathology , Encephalitis/diet therapy , Fatty Acids, Omega-3/administration & dosage , HMGB1 Protein/metabolism , Microglia/drug effects , Neuroprostanes/administration & dosage , Animals , Brain Edema/etiology , Brain Injuries, Traumatic/complications , Calcium-Binding Proteins/metabolism , Cerebral Cortex/pathology , Cytokines/metabolism , Disease Models, Animal , Encephalitis/etiology , Encephalitis/metabolism , Gene Expression Regulation/drug effects , Male , Microfilament Proteins/metabolism , Microglia/pathology , Nerve Tissue Proteins/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sirtuin 1/metabolism , Time Factors
7.
Age (Dordr) ; 38(5-6): 393-404, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27578256

ABSTRACT

High consumption of fruits and vegetables has been associated with reduced risk of debilitating diseases and improved cognition in aged populations. These beneficial effects have been attributed to the phytochemicals found in fruits and vegetables, which have previously been shown to be anti-inflammatory and modulate autophagy. Tart cherries contain a variety of potentially beneficial phytochemicals; however, little research has been done to investigate the effects of tart cherry on the aging brain. Therefore, the purpose of this study was to determine if tart cherry supplementation can improve cognitive and motor function of aged rats via modulation of inflammation and autophagy in the brain. Thirty 19-month-old male Fischer 344 rats were weight-matched and assigned to receive either a control diet or a diet supplemented with 2 % Montmorency tart cherry. After 6 weeks on the diet, rats were given a battery of behavioral tests to assess for strength, stamina, balance, and coordination, as well as learning and working memory. Although no significant effects were observed on tests of motor performance, tart cherry improved working memory of aged rats. Following behavioral testing, the hippocampus was collected for western/densitometric analysis of inflammatory (GFAP, NOX-2, and COX-2) and autophagy (phosphorylated mTOR, Beclin 1, and p62/SQSTM) markers. Tart cherry supplementation significantly reduced inflammatory markers and improved autophagy function. Daily consumption of tart cherry reduced age-associated inflammation and promoted protein/cellular homeostasis in the hippocampus, along with improvements in working memory. Therefore, addition of tart cherry to the diet may promote healthy aging and/or delay the onset of neurodegenerative diseases.


Subject(s)
Aging , Autophagy , Dietary Supplements , Encephalitis/diet therapy , Hippocampus/physiology , Memory, Short-Term , Prunus avium/chemistry , Animals , Behavior Rating Scale , Biomarkers/analysis , Cognition , Male , Maze Learning , Motor Activity , Powders , Rats , Rats, Inbred F344 , Time Factors
8.
J Nutr Biochem ; 26(11): 1166-73, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26315997

ABSTRACT

Neuroinflammation has been suggested as a central mediator of central nervous system dysfunction, including in dementia and neurodegenerative disease. Flavonoids have emerged as promising candidates for the prevention of neurodegenerative diseases and are thought to be capable of antiinflammatory effects in the brain. In the present study, the impact of a chronic intake of an anthocyanin extract from blackberry (BE) on brain inflammatory status in the presence or absence of a high-fat diet was investigated. Following intake of the dietary regimes for 17 weeks neuroinflammatory status in Wistar rat cortex, hippocampus and plasma were assessed using cytokine antibody arrays. In the cortex, intake of the high-fat diet resulted in an increase of at least 4-fold, in expression of the cytokine-induced neutrophil chemoattractant CINC-3, the ciliary neurotrophic factor CNTF, the platelet-derived growth factor PDGF-AA, IL-10, the tissue inhibitor of metalloproteinase TIMP-1 and the receptor for advanced glycation end products RAGE. BE intake partially decreased the expression of these mediators in the high-fat challenged brain. In standard-fed animals, BE intake significantly increased cortical levels of fractalkine, PDGF-AA, activin, the vascular endothelial growth factor VEGF and agrin expression, suggesting effects as neuronal growth and synaptic connection modulators. In hippocampus, BE modulates fractalkine and the thymus chemokine TCK-1 expression independently of diet intake and, only in standard diet, increased PDGF-AA. Exploring effects of anthocyanins on fractalkine transcription using the neuronal cell line SH-SY5Y suggested that other cell types may be involved in this effect. This is the first evidence, in in vivo model, that blackberry extract intake may be capable of preventing the detrimental effects of neuroinflammation in a high-fat challenged brain. Also, fractalkine and TCK-1 expression may be specific targets of anthocyanins and their metabolites on neuroinflammation.


Subject(s)
Diet, High-Fat/adverse effects , Inflammation/diet therapy , Neuroimmunomodulation/drug effects , Plant Extracts/pharmacology , Rubus , Animals , Anthocyanins/pharmacology , Body Weight/drug effects , Brain/drug effects , Brain/metabolism , Cell Line , Chemokine CX3CL1/genetics , Cytokines/metabolism , Encephalitis/diet therapy , Encephalitis/metabolism , Humans , Male , Microglia/drug effects , Plant Extracts/chemistry , Rats, Wistar , Rubus/chemistry
9.
Neurol Res ; 36(10): 906-10, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24735348

ABSTRACT

OBJECTIVE AND IMPORTANCE: Although ketogenic diet therapy is effective in refractory seizures in childhood, its effect on adult encephalitis with similar refractory seizures and prolonged encephalopathy has not been well reported. CLINICAL PRESENTATION: We report here a case of a 22-year-old man with acute encephalitis with refractory repetitive partial seizures (AERRPS). INTERVENTION: Partial seizures of the face developed to repeated generalized convulsions, which were refractory against anti-epileptic drugs and a high dose of propofol. After struggling for 9 months, he dramatically recovered after ketogenic diet therapy. CONCLUSION: Ketogenic diet therapy may be an important tool to help cure AERRPS.


Subject(s)
Diet, Ketogenic , Encephalitis/diet therapy , Encephalitis/physiopathology , Seizures/diet therapy , Seizures/physiopathology , Brain/pathology , Brain/physiopathology , Diffusion Magnetic Resonance Imaging , Drug Resistance , Electroencephalography , Encephalitis/pathology , Humans , Male , Seizures/drug therapy , Seizures/pathology , Young Adult
10.
Exp Neurol ; 232(2): 195-202, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21939657

ABSTRACT

Similar to fasting, the ketogenic diet (KD) has anti-inflammatory effects and protects against excitotoxicity-mediated neuronal cell death. Recent studies have shown that peroxisome proliferator-activated receptor (PPAR)γ has anti-inflammatory effects in seizure animal models. However, the exact mechanisms underlying the anti-inflammatory effects of the KD have not been determined for seizures. Here we investigated the effect of the KD and acetoacetate (AA) on neuroinflammation in a seizure animal model and glutamate-treated HT22 cells, respectively. Mice were fed the KD for 4 weeks and sacrificed 2 or 6h after KA injection. The KD reduced hippocampal tumor necrosis factor alpha (TNF-α) levels and nuclear factor (NF)-κB translocation into the nucleus 2h after KA treatment. KD-induced PPARγ activation was decreased by KA in neurons as assessed by western blotting and immunofluorescence. Finally, the KD inhibited cyclooxygenase (COX)-2 and microsomal prostaglandin E(2) synthase-1 (mPGES-1) expression in the hippocampus 6h after KA treatment. AA treatment also protected against glutamate-induced cell death in HT22 cells by reducing TNF-α and PPARγ-mediated COX-2 expression. Thus, the KD may inhibit neuroinflammation by suppressing a COX-2-dependent pathway via activation of PPARγ by the KD or AA.


Subject(s)
Diet, Ketogenic , Encephalitis/diet therapy , Epilepsy/chemically induced , Epilepsy/diet therapy , Hippocampus/immunology , PPAR gamma/metabolism , Acetoacetates/metabolism , Acetoacetates/pharmacology , Animals , Body Weight/physiology , CD11b Antigen/metabolism , Cell Line , Cell Survival/immunology , Cyclooxygenase 2/metabolism , Encephalitis/etiology , Encephalitis/immunology , Epilepsy/complications , Excitatory Amino Acid Agonists/toxicity , Glutamic Acid/toxicity , Hippocampus/cytology , Hippocampus/metabolism , Kainic Acid/toxicity , Male , Mice , Mice, Inbred ICR , Neurons/cytology , Neurons/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
J Neurochem ; 117(2): 264-74, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21272004

ABSTRACT

Glyceryl triacetate (GTA), a compound effective at increasing circulating and tissue levels of acetate was used to treat rats subjected to a continual 28 day intra-ventricular infusion of bacterial lipopolysaccharide (LPS). This model produces a neuroinflammatory injury characterized by global neuroglial activation and a decrease in choline acetyltransferase immunoreactivity in the basal forebrain. During the LPS infusion, rats were given a daily treatment of either water or GTA at a dose of 6 g/kg by oral gavage. In parallel experiments, free-CoA and acetyl-CoA levels were measured in microwave fixed brains and flash frozen heart, liver, kidney and muscle following a single oral dose of GTA. We found that a single oral dose of GTA significantly increased plasma acetate levels by 15 min and remained elevated for up to 4 h. At 30 min the acetyl-CoA levels in microwave-fixed brain and flash frozen heart and liver were increased at least 2.2-fold. The concentrations of brain acetyl-CoA was significantly increased between 30 and 45 min following treatment and remained elevated for up to 4 h. The concentration of free-CoA in brain was significantly decreased compared to controls at 240 min. Immunohistochemical and morphological analysis demonstrated that a daily treatment with GTA significantly reduced the percentage of reactive glial fibrillary acidic protein-positive astrocytes and activated CD11b-positive microglia by 40-50% in rats subjected to LPS-induced neuroinflammation. Further, in rats subjected to neuroinflammation, GTA significantly increased the number of choline acetyltransferase (ChAT)-positive cells by 40% in the basal forebrain compared to untreated controls. These data suggest that acetate supplementation increases intermediary short chain acetyl-CoA metabolism and that treatment is potentially anti-inflammatory and neuroprotective with regards to attenuating neuroglial activation and increasing ChAT immunoreactivity in this model.


Subject(s)
Acetates/pharmacology , Encephalitis/chemically induced , Encephalitis/diet therapy , Lipopolysaccharides , Acetates/blood , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , CD11b Antigen/metabolism , Cell Count/methods , Choline O-Acetyltransferase/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Encephalitis/pathology , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Male , Microglia/drug effects , Microglia/metabolism , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric , Time Factors
12.
Neurotox Res ; 15(1): 3-14, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19384583

ABSTRACT

Polyphenols extracted from grape seeds are able to inhibit amyloid-beta (Abeta) aggregation, reduce Abeta production and protect against Abeta neurotoxicity in vitro. We aimed to investigate the therapeutic effects of a polyphenol-rich grape seed extract (GSE) in Alzheimer's disease (AD) mice. APP(Swe)/PS1dE9 transgenic mice were fed with normal AIN-93G diet (control diet), AIN-93G diet with 0.07% curcumin or diet with 2% GSE beginning at 3 months of age for 9 months. Total phenolic content of GSE was 592.5 mg/g dry weight, including gallic acid (49 mg/g), catechin (41 mg/g), epicatechin (66 mg/g) and proanthocyanidins (436.6 mg catechin equivalents/g). Long-term feeding of GSE diet was well tolerated without fatality, behavioural abnormality, changes in food consumption, body weight or liver function. The Abeta levels in the brain and serum of the mice fed with GSE were reduced by 33% and 44%, respectively, compared with the Alzheimer's mice fed with the control diet. Amyloid plaques and microgliosis in the brain of Alzheimer's mice fed with GSE were also reduced by 49% and 70%, respectively. Curcumin also significantly reduced brain Abeta burden and microglia activation. Conclusively, polyphenol-rich GSE prevents the Abeta deposition and attenuates the inflammation in the brain of a transgenic mouse model, and this thus is promising in delaying development of AD.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Encephalitis/etiology , Grape Seed Extract/pharmacology , Age Factors , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/blood , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Body Weight/drug effects , Chromatography, High Pressure Liquid/methods , Cytokines/blood , Diet Therapy/methods , Disease Models, Animal , Encephalitis/diet therapy , Encephalitis/pathology , Enzyme-Linked Immunosorbent Assay , Flavonoids/pharmacology , Grape Seed Extract/chemistry , Humans , Liver/drug effects , Liver/enzymology , Mice , Mice, Transgenic , Peptide Fragments/blood , Phenols/pharmacology , Polyphenols , Presenilin-1/genetics
13.
J Neurol Sci ; 262(1-2): 27-36, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17681547

ABSTRACT

Alzheimer's disease (AD) is a major public health concern in all countries. Although the precise cause of AD is still unknown, a growing body of evidence supports the notion that soluble amyloid beta-peptide (Abeta) may be the proximate cause of synaptic injuries and neuronal death early in the disease. AD patients display lower levels of docosahexaenoic acid (DHA, C22:6 ; n-3) in plasma and brain tissues as compared to age-matched controls. Furthermore, epidemiological studies suggest that high DHA intake might have protective properties against neurodegenerative diseases. These observations are supported by in vivo studies showing that DHA-rich diets limits the synaptic loss and cognitive defects induced by Abeta peptide. Although the molecular basis of these neuroprotective effects remains unknown, several mechanisms have been proposed such as (i) regulation of the expression of potentially protective genes, (ii) activation of anti-inflammatory pathways, (iii) modulation of functional properties of the synaptic membranes along with changes in their physicochemical and structural features.


Subject(s)
Alzheimer Disease/diet therapy , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/antagonists & inhibitors , Brain/drug effects , Brain/metabolism , Docosahexaenoic Acids/therapeutic use , Encephalitis/diet therapy , Neuroprotective Agents/therapeutic use , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Brain/physiopathology , Docosahexaenoic Acids/metabolism , Encephalitis/metabolism , Encephalitis/physiopathology , Food, Formulated/standards , Humans , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Neuroprotective Agents/metabolism , Synapses/drug effects , Synapses/metabolism
14.
J Clin Neurosci ; 11(1): 66-7, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14642370

ABSTRACT

This case report describes the rare phenomenon of encephalopathy associated with massive carcinoid tumor. Extensive investigation failed to reveal an obvious cause but a presumptive diagnosis of tryptophan deficiency was made and she was commenced on tryptophan dietary supplements. A rapid and complete response resulted. This case report discusses this unusual case and reviews the literature regarding carcinoid associated encephalopathy.


Subject(s)
Antidepressive Agents, Second-Generation/administration & dosage , Carcinoid Tumor/diet therapy , Encephalitis/diet therapy , Tryptophan/administration & dosage , Adult , Carcinoid Tumor/complications , Carcinoid Tumor/pathology , Electroencephalography , Encephalitis/complications , Encephalitis/pathology , Female , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...