Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
Add more filters











Publication year range
1.
J Med Food ; 27(3): 242-249, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38354279

ABSTRACT

This study aimed to test the hypothesis that long-term and low-dose supplementation with an ethanol extract of Ecklonia stolonifera may confer protection against high-fat diet (HFD)-induced obesity in mice. Male C57BL/6J mice were divided into two groups, one of which was fed an HFD (40 kcal% fat) and the other an HFD+E. stolonifera (0.006%, w/w, ∼5 mg/kg body weight/day) for 16 weeks. E. stolonifera supplementation significantly reduced body weight from week 3 and until the end of the experiment. E. stolonifera-supplemented mice also exhibited lower fat mass (epididymal, perirenal, and mesenteric fat) and smaller adipocyte size than HFD control mice. The two groups displayed similar food intakes, but E. stolonifera markedly decreased lipogenesis and increased lipolysis and fatty acid oxidation in adipose tissue. Moreover, E. stolonifera significantly decreased plasma and hepatic lipid levels, hepatic lipid droplet accumulation, plasma aminotransferase levels, and liver weight by decreasing lipogenesis and increasing fatty acid oxidation. As E. stolonifera-supplemented mice showed improvements in hyperglycemia, insulin resistance, and inflammation, compared to control mice, it is possible that the beneficial effects of E. stolonifera on obesity might be associated with decreased inflammation and insulin resistance. Collectively, these results indicate that E. stolonifera could be used as a novel means of preventing and treating obesity and obesity-related metabolic disorders.


Subject(s)
Insulin Resistance , Non-alcoholic Fatty Liver Disease , Male , Animals , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Mice, Obese , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , Liver/metabolism , Obesity/drug therapy , Obesity/etiology , Inflammation/drug therapy , Plant Extracts/pharmacology , Fatty Acids/metabolism
2.
Antioxidants (Basel) ; 12(5)2023 May 08.
Article in English | MEDLINE | ID: mdl-37237929

ABSTRACT

Sarcopenic obesity, which refers to concurrent sarcopenia and obesity, is characterized by decreased muscle mass, strength, and performance along with abnormally excessive fat mass. Sarcopenic obesity has received considerable attention as a major health threat in older people. However, it has recently become a health problem in the general population. Sarcopenic obesity is a major risk factor for metabolic syndrome and other complications such as osteoarthritis, osteoporosis, liver disease, lung disease, renal disease, mental disease and functional disability. The pathogenesis of sarcopenic obesity is multifactorial and complicated, and it is caused by insulin resistance, inflammation, hormonal changes, decreased physical activity, poor diet and aging. Oxidative stress is a core mechanism underlying sarcopenic obesity. Some evidence indicates a protective role of antioxidant flavonoids in sarcopenic obesity, although the precise mechanisms remain unclear. This review summarizes the general characteristics and pathophysiology of sarcopenic obesity and focuses on the role of oxidative stress in sarcopenic obesity. The potential benefits of flavonoids in sarcopenic obesity have also been discussed.

3.
Biomedicines ; 11(5)2023 May 07.
Article in English | MEDLINE | ID: mdl-37239054

ABSTRACT

The blood-brain barrier (BBB) restricts entry of neurotoxic plasma components, blood cells, and pathogens into the brain, leading to proper neuronal functioning. BBB impairment leads to blood-borne protein infiltration such as prothrombin, thrombin, prothrombin kringle-2, fibrinogen, fibrin, and other harmful substances. Thus, microglial activation and release of pro-inflammatory mediators commence, resulting in neuronal damage and leading to impaired cognition via neuroinflammatory responses, which are important features observed in the brain of Alzheimer's disease (AD) patients. Moreover, these blood-borne proteins cluster with the amyloid beta plaque in the brain, exacerbating microglial activation, neuroinflammation, tau phosphorylation, and oxidative stress. These mechanisms work in concert and reinforce each other, contributing to the typical pathological changes in AD in the brain. Therefore, the identification of blood-borne proteins and the mechanisms involved in microglial activation and neuroinflammatory damage can be a promising therapeutic strategy for AD prevention. In this article, we review the current knowledge regarding the mechanisms of microglial activation-mediated neuroinflammation caused by the influx of blood-borne proteins into the brain via BBB disruption. Subsequently, the mechanisms of drugs that inhibit blood-borne proteins, as a potential therapeutic approach for AD, along with the limitations and potential challenges of these approaches, are also summarized.

4.
J Clin Med ; 12(5)2023 Feb 22.
Article in English | MEDLINE | ID: mdl-36902541

ABSTRACT

This study investigated the therapeutic effects of transplanting human mesenchymal stem cells (hMSCs) into wild-type mice that were intraperitoneally administered cytosine arabinoside (Ara-C) to develop cerebellar ataxia (CA) during the first three postnatal days. hMSCs were intrathecally injected into 10-week-old mice once or thrice at 4-week intervals. Compared to the nontreated mice, the hMSC-treated mice showed improved motor and balance coordination, as measured using the rotarod, open-field, and ataxic scoring assessments, and increased protein levels in Purkinje and cerebellar granule cells, as measured using calbindin and NeuN protein markers. Multiple hMSC injections preserved Ara-C-induced cerebellar neuronal loss and improved cerebellar weight. Furthermore, the hMSC implantation significantly elevated the levels of neurotrophic factors, including brain-derived and glial cell line-derived neurotrophic factors, and suppressed TNF-α-, IL-1ß-, and iNOS-mediated proinflammatory responses. Collectively, our results demonstrate that hMSCs exhibit therapeutic potential for Ara-C-induced CA by protecting neurons through the stimulation of neurotrophic factors and inhibition of cerebellar inflammatory responses, which can improve motor behavior and alleviate ataxia-related neuropathology. In summary, this study suggests that hMSC administration, particularly multiple treatments, can effectively treat ataxia-related symptoms with cerebellar toxicity.

5.
J Food Biochem ; 46(12): e14501, 2022 12.
Article in English | MEDLINE | ID: mdl-36332134

ABSTRACT

Lignan schisandrol A (SolA) is known to have antioxidant and anti-inflammatory effects. However, the impact of SolA on obesity is poorly understood. To test the hypothesis that SolA has anti-obesity effects, C57BL/6J mice were fed a high-fat diet with or without SolA (0.006%, w/w) for 16 weeks. SolA decreased visceral fat mass (10%) by increasing energy expenditure and upregulating white adipose tissue thermogenic genes mRNA expression. Furthermore, SolA upregulated adipose Lpl mRNA expression and decreased plasma free fatty acid (FFA), triglyceride (TG), apolipoprotein (apo) B, aspartate aminotransferase levels and TG/HDL-cholesterol and apoB/apoA1 ratios as well as hepatic lipid droplets. Increased hepatic ß-oxidation and fecal FFA and TG levels were observed in the SolA-supplemented mice, suggesting an association of its lipid-lowering effect with increased hepatic ß-oxidation, fecal fat excretion and adipose Lpl. Conclusionally, this study provides evidence on the protective effects of SolA against adiposity, dyslipidemia and nonalcoholic fatty liver disease in obese mice.


Subject(s)
Lignans , Obesity , Animals , Mice , Mice, Obese , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/etiology , Lignans/pharmacology , Diet, High-Fat/adverse effects , RNA, Messenger/metabolism
6.
Prev Nutr Food Sci ; 27(4): 407-413, 2022 Dec 31.
Article in English | MEDLINE | ID: mdl-36721747

ABSTRACT

Sinapic acid (SA), a hydroxycinnamic acid, is known to confer protection against oxidative stress, inflammation, diabetes, and liver disease. However, the effectiveness of SA in improving obesity remains obscure. Therefore, this study evaluated anti-obesity efficacy of SA and to elucidate its mechanism of action. Male mice were maintained for 16 weeks on high-fat diet (HFD) alone or with SA (0.004%, w/w) and bodyweight, fat mass, adipocyte size, food intake, and biochemical and molecular markers were evaluated. SA-supplemented mice demonstrated markedly decreased fat mass and adipocyte size compared to unsupplemented group, without any changes in bodyweight and food intake between the two groups. Plasma adipocytokines levels including leptin, resistin, monocyte chemoattractant protein (MCP)-1 and interleukin-6 were also markedly reduced by SA supplementation. SA tended to lower plasma insulin level and improved homeostatic index of insulin resistance and intraperitoneal glucose tolerance test in HFD-induced obese mice. The anti-adiposity effect of SA was maybe owing to down-regulation of the mRNA expression of lipogenic genes, including acetyl coenzyme A (CoA) carboxylase, fatty acid synthesis, stearoyl-CoA desaturase 1, and phosphatidate phosphatase, and peroxisome proliferator-activated receptor γ, a transcription factor responsible for governing lipid metabolism, in adipose tissues. SA significantly down-regulated pro-inflammatory nuclear factor kappa B, MCP-1, tumor necrosis factor-α, and Toll-like receptor 4 mRNA expression in adipose tissue. Thus, SA could be beneficial for the development of functional foods or herbal medications to combat obesity.

7.
Nutr Res Pract ; 15(4): 431-443, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34349877

ABSTRACT

BACKGROUND/OBJECTIVES: Nobiletin (NOB), a citrus flavonoid, is reported to have beneficial effects on cardiovascular and metabolic health. However, there is limited research investigating the effect of long-term supplementation with low-dose NOB on high-cholesterol diet (HCD)-induced hypercholesterolemia and non-obese nonalcoholic fatty liver disease (NAFLD). Therefore, we investigated the influence of NOB on hypercholesterolemia and NAFLD in HCD-fed mice. SUBJECTS/METHODS: C57BL/6J mice were fed a normal diet (ND) or HCD (35 kcal% fat, 1.25% cholesterol, 0.5% cholic acid) with or without NOB (0.02%) for 20 weeks. RESULTS: HCD feeding markedly reduced the final body weight compared to ND feeding, with no apparent energy intake differences. NOB supplementation suppressed HCD-induced weight loss without altering energy intake. Moreover, NOB significantly decreased the total cholesterol (TC) levels and the low-density lipoprotein (LDL)/very-LDL-cholesterol to TC ratio, and increased the high-density lipoprotein-cholesterol/TC ratio in plasma, compared to those for HCD feeding alone. The plasma levels of inflammatory and atherosclerosis markers (C-reactive protein, oxidized LDL, interleukin [IL]-1ß, IL-6, and plasminogen activator inhibitor-1) were significantly lower, whereas those of anti-atherogenic adiponectin and paraoxonase were higher in the NOB-supplemented group than in the HCD control group. Furthermore, NOB significantly decreased liver weight, hepatic cholesterol and triglyceride contents, and lipid droplet accumulation by inhibiting messenger RNA expression of hepatic genes and activity levels of cholesterol synthesis-, esterification-, and fatty acid synthesis-associated enzymes, concomitantly enhancing fatty acid oxidation-related gene expression and enzyme activities. Dietary NOB supplementation may protect against hypercholesterolemia and NAFLD via regulation of hepatic lipid metabolism in HCD-fed mice; these effects are associated with the amelioration of inflammation and reductions in the levels of atherosclerosis-associated cardiovascular markers. CONCLUSIONS: The present study suggests that NOB may serve as a potential therapeutic agent for the treatment of HCD-induced hypercholesterolemia and NAFLD.

8.
Biomed Pharmacother ; 142: 111969, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34333285

ABSTRACT

p-Coumaric acid (PC), a naturally occurring phytochemical, possesses antioxidant and anti-inflammatory properties; however, the mechanisms underlying its protective effects against obesity-related metabolic dysfunction are largely unknown. Here, we treated C57BL/6J mice to a high-fat diet (HFD) with or without PC (10 mg/kg body weight/day) for 16 weeks to determine whether PC ameliorates HFD-induced obesity, insulin resistance, inflammation, and non-alcoholic fatty liver disease (NAFLD). We found no significant differences in food intake and body weight between the groups. However, PC-treated mice showed significantly lower white adipose tissue (WAT) weight, adipocyte size, and plasma leptin level, which were associated with decreased lipogenic enzyme activity and mRNA expression of their genes in the epididymal WAT. Moreover, hepatic lipogenic enzymes activities and expression of their genes and proteins were decreased with concomitant increases in hepatic fatty acid oxidation and mRNA expression of its gene; fecal lipid excretion was significantly increased, resulting in decreased liver weight, hepatic lipid levels, lipid droplet accumulation, and plasma aspartate aminotransferase and lipid levels. Additionally, PC-treated mice showed lower fasting blood glucose, plasma resistin, and MCP-1 levels, HOMA-IR, and mRNA expression of inflammatory genes in the epididymal WAT and liver. Our findings reveal potential mechanisms underlying the action of PC against HFD-induced adiposity, NAFLD, and other metabolic disturbances.


Subject(s)
Coumaric Acids/pharmacology , Inflammation/prevention & control , Non-alcoholic Fatty Liver Disease/prevention & control , Obesity/prevention & control , Adiposity/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Blood Glucose/drug effects , Diet, High-Fat/adverse effects , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Resistin/blood
9.
Int J Mol Sci ; 22(9)2021 May 03.
Article in English | MEDLINE | ID: mdl-34063708

ABSTRACT

Alzheimer's disease (AD) is the most frequent cause of age-related neurodegeneration and cognitive impairment, and there are currently no broadly effective therapies. The underlying pathogenesis is complex, but a growing body of evidence implicates mitochondrial dysfunction as a common pathomechanism involved in many of the hallmark features of the AD brain, such as formation of amyloid-beta (Aß) aggregates (amyloid plaques), neurofibrillary tangles, cholinergic system dysfunction, impaired synaptic transmission and plasticity, oxidative stress, and neuroinflammation, that lead to neurodegeneration and cognitive dysfunction. Indeed, mitochondrial dysfunction concomitant with progressive accumulation of mitochondrial Aß is an early event in AD pathogenesis. Healthy mitochondria are critical for providing sufficient energy to maintain endogenous neuroprotective and reparative mechanisms, while disturbances in mitochondrial function, motility, fission, and fusion lead to neuronal malfunction and degeneration associated with excess free radical production and reduced intracellular calcium buffering. In addition, mitochondrial dysfunction can contribute to amyloid-ß precursor protein (APP) expression and misprocessing to produce pathogenic fragments (e.g., Aß1-40). Given this background, we present an overview of the importance of mitochondria for maintenance of neuronal function and how mitochondrial dysfunction acts as a driver of cognitive impairment in AD. Additionally, we provide a brief summary of possible treatments targeting mitochondrial dysfunction as therapeutic approaches for AD.


Subject(s)
Alzheimer Disease/genetics , Cognitive Dysfunction/genetics , Oxidative Stress/genetics , Plaque, Amyloid/genetics , Alzheimer Disease/pathology , Brain/metabolism , Brain/pathology , Cognitive Dysfunction/pathology , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Neurons/metabolism , Neurons/pathology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
10.
Adv Nutr ; 11(3): 742, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32419021

Subject(s)
Epilepsy , Flavonoids , Humans
11.
Int J Mol Sci ; 21(5)2020 Mar 09.
Article in English | MEDLINE | ID: mdl-32182914

ABSTRACT

To test the hypothesis that myricitrin (MYR) improves type 2 diabetes, we examined the effect of MYR on hyperglycemia, glucose intolerance, hepatic steatosis, and inflammation in high-fat diet (HFD) and streptozotocin (STZ)-induced type 2 diabetic mice. Male C57BL/6J mice were randomly divided into three groups: non-diabetic, diabetic control, and MYR (0.005%, w/w)-supplemented diabetic groups. Diabetes was induced by HFD and STZ, and MYR was administered orally for 5 weeks. Myricitrin exerted no significant effects on food intake, body weight, fat weight, or plasma lipids levels. However, MYR significantly decreased fasting blood glucose levels, improved glucose intolerance, and increased pancreatic ß-cell mass compared to the diabetic control group. Myricitrin administration also markedly increased glucokinase mRNA expression and activity as well as lowered glucose-6-phosphatase and phosphoenolpyruvate carboxykinase mRNA expression and activity in the liver. In addition, liver weight, hepatic triglyceride content, and lipid droplet accumulation were markedly decreased following MYR administration. These changes were seemingly attributable to the suppression of the hepatic lipogenic enzymes-fatty acid synthase and phosphatidate phosphohydrolase. Myricitrin also significantly lowered plasma MCP-1 and TNF-α levels and the mRNA expression of hepatic pro-inflammatory genes. These results suggest that MYR has anti-diabetic potential.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Fatty Liver/drug therapy , Flavonoids/pharmacology , Glucose Intolerance/drug therapy , Hyperglycemia/drug therapy , Inflammation/drug therapy , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/drug therapy , Diet, High-Fat/adverse effects , Fatty Liver/metabolism , Glucokinase/metabolism , Glucose Intolerance/metabolism , Hyperglycemia/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Insulin Resistance , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Streptozocin/pharmacology , Triglycerides/metabolism
12.
J Med Food ; 23(2): 198-202, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31913760

ABSTRACT

Temporal lobe epilepsy (TLE) is the most common form of localization-related epilepsy, with the highest prevalence rate in adulthood. Recently, we reported the beneficial effects of the individual treatment with flavonoids such as silibinin and morin in kainic acid (KA)-treated mouse model for TLE. In this study, we investigated whether there is a synergistic effect of co-treatment with silibinin and morin on the susceptibility to seizure, the frequency of spontaneous recurrent seizures (SRSs), and granule cell dispersion in the dentate gyrus, which could be partially controlled by treatment with each flavonoid in the animal model for TLE. Unfortunately, we did not observe any synergistic effect against the susceptibility of seizure and SRS induced by KA treatment. However, the combination of these flavonoids showed similar antiepileptic effects compared with treatment with each one individually. Therefore, although silibinin and morin are not suitable for combination therapy, our results still suggest that these flavonoids can be used as potent therapeutic compounds for preventing epileptic seizures.


Subject(s)
Epilepsy, Temporal Lobe/drug therapy , Flavonoids/therapeutic use , Seizures/drug therapy , Silybin/therapeutic use , Animals , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Drug Synergism , Epilepsy, Temporal Lobe/chemically induced , Kainic Acid , Mice , Seizures/chemically induced
13.
J Med Food ; 22(11): 1151-1158, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31549892

ABSTRACT

The flavonoid myricitrin exhibits various pharmacological and physiological effects. However, studies on the effects of myricitrin on obesity are limited. We hypothesized that dietary myricitrin would attenuate the adiposity and metabolic dysfunction that occur in obesity. To test this hypothesis, mice were randomly fed a high-fat diet (HFD) or HFD supplemented with myricitrin for 16 weeks. Myricitrin significantly reduced white adipose tissue (WAT) mass, adipocyte size, and plasma leptin levels, and also attenuated dyslipidemia. These changes appeared to result from increased energy expenditure and activation of the carnitine acyltransferase (CPT) and ß-oxidation in WAT. Expressions of the proinflammatory genes NF-κB, TLR2, MCP1, and TNF-α were also lower in the WAT of myricitrin-supplemented mice. Moreover, myricitrin markedly reduced hepatic triglyceride accumulation and plasma aspartate transaminase levels by increasing CPT activity and reducing fatty acid synthase activity in the liver. Myricitrin-supplemented mice also showed improved glucose tolerance, insulin sensitivity, and decreased hyperinsulinemia, along with decreased levels of circulating resistin. In conclusion, long-term consumption of a myricitrin-supplemented diet may effectively protect against HFD-induced obesity and related metabolic disorders.


Subject(s)
Adiposity , Dietary Supplements , Flavonoids/pharmacology , Obesity/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Diet, High-Fat/adverse effects , Dyslipidemias/prevention & control , Fatty Liver/prevention & control , Inflammation/prevention & control , Insulin Resistance , Leptin/blood , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Obesity/chemically induced
14.
Adv Nutr ; 10(5): 778-790, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31111873

ABSTRACT

Epilepsy is a chronic neurological disorder that affects many people worldwide. Temporal lobe epilepsy is the most common and most studied type of epilepsy, but the pathological mechanisms underlying this condition are poorly understood. More than 20 antiepileptic drugs (AEDs) have been developed and used for the treatment of epilepsy; however, 30% of patients still experience uncontrolled epilepsy and associated comorbidities, which impair their quality of life. In addition, various side effects have been reported for AEDs, such as drowsiness, unsteadiness, dizziness, blurred or double vision, tremor (shakiness), greater risk of infections, bruising, and bleeding. Thus, critical medical needs remain unmet for patients with uncontrolled epilepsy. Flavonoids belong to a subclass of polyphenols that are widely present in fruits, vegetables, and certain beverages. Recently, many studies have reported that some flavonoids elicit various beneficial effects in patients with epilepsy without causing the side effects associated with conventional medical therapies. Moreover, flavonoids may have a property of regulating microRNA expression associated with inflammation and cell survival. These findings suggest that flavonoids, which are more effective but impose fewer adverse effects than conventional AEDs, could be used in the treatment of epilepsy.


Subject(s)
Anticonvulsants/therapeutic use , Complementary Therapies/methods , Epilepsy/therapy , Flavonoids/therapeutic use , Humans
15.
Int J Mol Sci ; 20(9)2019 May 09.
Article in English | MEDLINE | ID: mdl-31075962

ABSTRACT

This study focuses on the effect of honokiol (HON) on glucose homeostasis, insulin resistance, dyslipidemia, hepatic steatosis, and inflammation in type 2 diabetic db/db mice. Male C57BL/KsJ-db/db mice were fed a normal diet with or without HON (0.02%, w/w) or pioglitazone (PIO, anti-diabetic agent, 0.01%, w/w) for 5 weeks. Blood biomarker, tissue morphology and enzymatic and genetic parameters were determined. PIO significantly decreased food intake, fasting blood glucose, and glycosylated hemoglobin (HbA1c) levels, but markedly increased body weight, adipose tissue weight, and plasma leptin levels. HON did not significantly affect food intake, body weight, or levels of plasma leptin and blood glucose. However, HON led to significant decreases in adipose tissue weight, plasma insulin, blood HbA1c and HOMA-IR levels and improved glucose tolerance. The anti-diabetic and anti-adiposity effects of HON were partially related to the inhibition of gluconeogenic enzymes and their mRNA expression in the liver; and the inhibition of lipogenic enzymes in adipose tissue, respectively. Unlike PIO, HON did not affect dyslipidemia, but ameliorated hepatic steatosis by inhibiting hepatic lipogenic enzymes activity. Moreover, HON exhibited anti-inflammatory effects similar to PIO. These results suggest that HON can protect against type 2 diabetes by improving insulin resistance, glucose and lipid metabolism, and inflammation.


Subject(s)
Biphenyl Compounds/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Fatty Liver/drug therapy , Inflammation/drug therapy , Insulin Resistance , Lignans/therapeutic use , Adipose Tissue/drug effects , Adipose Tissue/pathology , Animals , Biphenyl Compounds/pharmacology , Blood Glucose/metabolism , Body Weight/drug effects , Cytokines/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/pathology , Dyslipidemias/blood , Dyslipidemias/complications , Fatty Liver/blood , Fatty Liver/complications , Fatty Liver/pathology , Feeding Behavior/drug effects , Homeostasis/drug effects , Inflammation/blood , Inflammation/complications , Inflammation/pathology , Inflammation Mediators/metabolism , Leptin/blood , Lignans/pharmacology , Lipids/blood , Male , Mice, Inbred C57BL , Organ Size/drug effects
16.
Nutr Res ; 64: 64-71, 2019 04.
Article in English | MEDLINE | ID: mdl-30802724

ABSTRACT

We hypothesized that schizandrin (SCH) A, a lignan found in the fruits of the Schisandra genus, would exert protective effects against high-fat and high-cholesterol (HFHC) diet-induced nonalcoholic fatty liver disease (NAFLD) via regulation of lipid metabolism and oxidative stress. To test our hypothesis, male C57BL/6J mice were fed an HFHC diet with or without SCH A for 15 weeks. There were no significant differences in food intake, body weight, fat mass, and plasma total cholesterol level between the 2 groups. However, supplementation of SCH A significantly decreased levels of plasma free fatty acid and triglyceride, whereas plasma high-density lipoprotein cholesterol level was increased in the SCH A-supplemented mice. Moreover, hepatic free fatty acid, triglyceride, and cholesterol content, as well as hepatic lipid droplet accumulation, were markedly lower in the SCH A group in contrast to the control group. Activity of hepatic enzymes involved in fatty acid and triglyceride synthesis was significantly decreased by SCH A supplementation, whereas SCH A markedly increased hepatic ß-oxidation and fatty acid oxidation-related gene expression as well as fecal excretion of free fatty acid and triglyceride. SCH A also significantly increased expression of genes involved in cholesterol homeostasis (biliary cholesterol excretion and cholesterol efflux to high-density lipoprotein) in the liver. Moreover, SCH A significantly decreased hepatic lipid peroxidation, which was accompanied by increased hepatic antioxidant enzymes activity. These results suggest that SCH A could alleviate HFHC diet-induced NAFLD by regulating hepatic lipid metabolism and oxidative stress as well as fecal lipid excretion.


Subject(s)
Cholesterol, Dietary/metabolism , Cyclooctanes/therapeutic use , Lignans/therapeutic use , Lipid Metabolism/drug effects , Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Oxidative Stress/drug effects , Polycyclic Compounds/therapeutic use , Schisandra/chemistry , Animals , Antioxidants/metabolism , Cholesterol/blood , Cholesterol/metabolism , Cholesterol, Dietary/administration & dosage , Cholesterol, Dietary/adverse effects , Cholesterol, Dietary/blood , Cholesterol, HDL/blood , Cyclooctanes/pharmacology , Diet, High-Fat , Dietary Fats/administration & dosage , Dietary Fats/adverse effects , Dietary Fats/metabolism , Dietary Supplements , Fatty Acids/metabolism , Fatty Acids, Nonesterified/blood , Feces/chemistry , Lignans/pharmacology , Liver/metabolism , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Phytotherapy , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Polycyclic Compounds/pharmacology , Triglycerides/blood
17.
J Med Food ; 22(3): 277-285, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30632945

ABSTRACT

Parkinson's disease (PD) and Alzheimer's disease exhibit common features of neurodegenerative diseases and can be caused by numerous factors. A common feature of these diseases is neurotoxic inflammation by activated microglia, indicating that regulation of microglial activation is a potential mechanism for preserving neurons in the adult brain. Recently, we reported that upregulation of prothrombin kringle-2 (pKr-2), one of the domains that make up prothrombin and which is cleaved and generated by active thrombin, induces nigral dopaminergic (DA) neuronal death through neurotoxic microglial activation in the adult brain. In this study, we show that silibinin, a flavonoid found in milk thistle, can suppress the production of inducible nitric oxide synthase and neurotoxic inflammatory cytokines, such as interleukin-1ß and tumor necrosis factor-α, after pKr-2 treatment by downregulating the extracellular signal-regulated kinase signaling pathway in the mouse substantia nigra. Moreover, as demonstrated by immunohistochemical staining, measurements of the dopamine and metabolite levels, and open-field behavioral tests, silibinin treatment protected the nigrostriatal DA system resulting from the occurrence of pKr-2-triggered neurotoxic inflammation in vivo. Thus, we conclude that silibinin may be beneficial as a natural compound with anti-inflammatory effects against pKr-2-triggered neurotoxicity to protect the nigrostriatal DA pathway and its properties, and thus, may be applicable for PD therapy.


Subject(s)
Dopamine/metabolism , Parkinson Disease/drug therapy , Prothrombin/toxicity , Silybin/administration & dosage , Animals , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Kringles , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Parkinson Disease/etiology , Parkinson Disease/metabolism , Prothrombin/chemistry , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
18.
J Med Food ; 21(12): 1306-1309, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30136878

ABSTRACT

Abnormal reorganization of the dentate gyrus and neuroinflammation in the hippocampus represent characteristic phenotypes of patients suffering from temporal lobe epilepsy. Hesperetin, a flavanone abundant in citrus fruit, is known to have protective effects by preventing inflammation and oxidative stress in neuronal cultures and in the adult murine brain. However, the protective effects of hesperetin against epileptic seizures in vivo remain unclear, despite one study reporting anticonvulsant effects in vitro. In this study, we report that oral administration of hesperetin not only delays the onset of seizures triggered by kainic acid (KA) but also contributes to the attenuation of granule cell dispersion in the KA-treated hippocampus. Moreover, we observed that hesperetin administration inhibited the expression of pro-inflammatory molecules produced by activated microglia in the hippocampus. Thus, administration of hesperetin might be beneficial for preventing epileptic seizures.


Subject(s)
Anticonvulsants/therapeutic use , Citrus , Epilepsy, Temporal Lobe/drug therapy , Hesperidin/therapeutic use , Phytotherapy , Administration, Oral , Animals , Anticonvulsants/administration & dosage , Disease Models, Animal , Epilepsy, Temporal Lobe/chemically induced , Fruit , Hesperidin/administration & dosage , Kainic Acid , Male , Mice
19.
Exp Neurobiol ; 27(3): 226-237, 2018 Jun.
Article in English | MEDLINE | ID: mdl-30022874

ABSTRACT

An abnormal reorganization of the dentate gyrus and neurotoxic events are important phenotypes in the hippocampus of patients with temporal lobe epilepsy (TLE). The effects of morin, a bioflavonoid constituent of many herbs and fruits, on epileptic seizures have not yet been elucidated, though its beneficial effects, such as its anti-inflammatory and neuroprotective properties, are well-described in various neurodegenerative diseases. In the present study, we investigated whether treatment with morin hydrate (MH) can reduce the susceptibility to seizures, granule cell dispersion (GCD), mammalian target of rapamycin complex 1 (mTORC1) activity, and the increases in the levels of apoptotic molecules and inflammatory cytokines in the kainic acid (KA)-induced seizure mouse model. Our results showed that oral administration of MH could reduce susceptibility to seizures and lead to the inhibition of GCD and mTORC1 activity in the KA-treated hippocampus. Moreover, treatment with MH significantly reduced the increased levels of apoptotic signaling molecules and pro-inflammatory mediators in the KA-treated hippocampus compared with control mice, suggesting a neuroprotective role. Therefore, these results suggest that morin has a therapeutic potential against epilepsy through its abilities to inhibit GCD and neurotoxic events in the in vivo hippocampus.

20.
Cell Death Dis ; 9(5): 449, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29670079

ABSTRACT

The role of astrocyte elevated gene-1 (AEG-1) in nigral dopaminergic (DA) neurons has not been studied. Here we report that the expression of AEG-1 was significantly lower in DA neurons in the postmortem substantia nigra of patients with Parkinson's disease (PD) compared to age-matched controls. Similarly, decreased AEG-1 levels were found in the 6-hydroxydopamine (6-OHDA) mouse model of PD. An adeno-associated virus-induced increase in the expression of AEG-1 attenuated the 6-OHDA-triggered apoptotic death of nigral DA neurons. Moreover, the neuroprotection conferred by the AEG-1 upregulation significantly intensified the neurorestorative effects of the constitutively active ras homolog enriched in the brain [Rheb(S16H)]. Collectively, these results demonstrated that the sustained level of AEG-1 as an important anti-apoptotic factor in nigral DA neurons might potentiate the therapeutic effects of treatments, such as Rheb(S16H) administration, on the degeneration of the DA pathway that characterizes PD.


Subject(s)
Apoptosis , Astrocytes/metabolism , Dopaminergic Neurons/metabolism , Membrane Glycoproteins/biosynthesis , Substantia Nigra/metabolism , Up-Regulation , Animals , Astrocytes/pathology , Disease Models, Animal , Dopaminergic Neurons/pathology , Humans , Membrane Glycoproteins/genetics , Mice , Oxidopamine/adverse effects , Oxidopamine/pharmacology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/genetics , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , Ras Homolog Enriched in Brain Protein/genetics , Ras Homolog Enriched in Brain Protein/metabolism , Substantia Nigra/pathology
SELECTION OF CITATIONS
SEARCH DETAIL