Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 234
Filter
1.
J Med Food ; 24(8): 873-882, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34406876

ABSTRACT

Ancient traditions showed that fermented enzyme foods have beneficial health effects on the body. However, only a few studies have reported on its impact on weight loss and metabolic syndrome. Therefore, it is necessary to verify whether diet supplementation with fermented enzyme foods can have a beneficial functional impact on the body. We examined the antiobesity properties of fermented mixed grain (FMG) with digestive enzymes (FMG) in diet-induced obese mice. Sixty C57BL/6J mice were randomly assigned to six dietary groups: (1) normal diet (ND), (2) high-fat diet (HFD), (3) Bacilus Coagulans, (4) steamed grain, (5) low-dose FMG (L-FMG), and (6) high-dose FMG (H-FMG) supplement for 12 weeks. The results showed that H-FMG supplement dramatically decreased body weight and fat mass with simultaneous decreases in plasma lipid contents. Furthermore, H-FMG significantly lowered fasting blood glucose concentrations and improved glucose tolerance compared with the HFD group. Also, the concentrations of inflammatory cytokines secreted from adipocytes in H-FMG-supplemented mice decreased dramatically. Taken together, our findings indicated that H-FMG can ameliorate HFD-induced obesity and its associated complications and could be used as a potential preventive intervention for obesity.


Subject(s)
Diet, High-Fat , Metabolic Diseases , Adiposity , Animals , Body Weight , Diet, High-Fat/adverse effects , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity
2.
Int J Mol Sci ; 22(1)2020 Dec 28.
Article in English | MEDLINE | ID: mdl-33379198

ABSTRACT

Obesity, a characteristic of metabolic syndrome, is also associated with chronic inflammation and the development of autoimmune diseases. However, the relationship between obesity and autoimmune diseases remains to be investigated in depth. Here, we compared hepatic gene expression profiles among high-fat diet (HFD) mice using the primary biliary cholangitis (PBC) mouse model based on the chronic expression of interferon gamma (IFNγ) (ARE-Del-/- mice). The top differentially expressed genes affected by upstream transcriptional regulators IFNγ, LPS, and TNFα displayed an overlap in HFD and ARE-Del-/- mice, indicating that obesity-induced liver inflammation may be dependent on signaling via IFNγ. The top pathways altered in HFD mice were mostly involved in the innate immune responses, which overlapped with ARE-Del-/- mice. In contrast, T cell-mediated signaling pathways were exclusively altered in ARE-Del-/- mice. We further evaluated the therapeutic effect of luteolin, known as anti-inflammatory flavonoid, in HFD and ARE-Del-/- mice. Luteolin strongly suppressed the MHC I and II antigen presentation pathways, which were highly activated in both HFD and ARE-Del-/- mice. Conversely, luteolin increased metabolic processes of fatty acid oxidation and oxidative phosphorylation in the liver, which were suppressed in ARE-Del-/- mice. Luteolin also strongly induced PPAR signaling, which was downregulated in HFD and ARE-Del-/- mice. Using human GWAS data, we characterized the genetic interaction between significant obesity-related genes and IFNγ signaling and demonstrated that IFNγ is crucial for obesity-mediated inflammatory responses. Collectively, this study improves our mechanistic understanding of the relationship between obesity and autoimmune diseases. Furthermore, it provides new methodological insights into how immune network-based analyses effectively integrate RNA-seq and microarray data.


Subject(s)
Diet, High-Fat/adverse effects , Interferon-gamma/metabolism , Leptin/metabolism , Liver Cirrhosis, Biliary/etiology , Obesity/immunology , Animals , Antigen Presentation/drug effects , Energy Metabolism/drug effects , Liver/metabolism , Liver Cirrhosis, Biliary/prevention & control , Luteolin/pharmacology , Luteolin/therapeutic use , Mice , Obesity/complications , Obesity/genetics , Obesity/metabolism , Signal Transduction/drug effects , Th1 Cells
3.
Nutrients ; 12(10)2020 Sep 29.
Article in English | MEDLINE | ID: mdl-33003339

ABSTRACT

The Chrysanthemum morifolium Ramat (CM) is widely used as a traditional medicine and herbal tea by the Asian population for its health benefits related to obesity. However, compared to the flowers of CM, detailed mechanisms underlying the beneficial effects of its leaves on obesity and dyslipidemia have not yet been elucidated. Therefore, to investigate the lipidomic biomarkers responsible for the pharmacological effects of CM leaf extract (CLE) in plasma of mice fed a high-fat diet (HFD), the plasma of mice fed a normal diet (ND), HFD, HFD plus CLE 1.5% diet, and HFD plus luteolin 0.003% diet (LU) for 16 weeks were analyzed using liquid chromatography-tandem mass spectrometry (LC-MS/MS) combined with multivariate analysis. In our analysis, the ND, HFD, CLE, and LU groups were clearly differentiated by partial least-squares discriminant analysis (PLS-DA) score plots. The major metabolites contributing to this differentiation were cholesteryl esters (CEs), lysophosphatidylcholines (LPCs), phosphatidylcholines (PCs), ceramides (CERs), and sphingomyelins (SMs). The levels of plasma CEs, LPCs, PCs, SMs, and CERs were significantly increased in the HFD group compared to those in the ND group, and levels of these lipids recovered to normal after administration of CLE or LU. Furthermore, changes in hepatic mRNA expression levels involved in the Kennedy pathway and sphingolipid biosynthesis were also suppressed by treatment with CLE or LU. In conclusion, this study examined the beneficial effects of CLE and LU on obesity and dyslipidemia, which were demonstrated as reduced synthesis of lipotoxic intermediates. These results may provide valuable insights towards evaluating the therapeutic effects of CLE and LU and understanding obesity-related diseases.


Subject(s)
Anti-Obesity Agents/pharmacology , Chrysanthemum , Dyslipidemias/blood , Obesity/blood , Plant Extracts/pharmacology , Animals , Ceramides/blood , Cholesterol Esters/blood , Chromatography, Liquid , Diet, High-Fat/adverse effects , Dietary Supplements , Dyslipidemias/etiology , Dyslipidemias/therapy , Lipidomics , Liver/metabolism , Luteolin/pharmacology , Lysophosphatidylcholines/blood , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/therapy , Phosphatidylcholines/blood , Plant Leaves , RNA, Messenger/metabolism , Sphingomyelins/blood , Tandem Mass Spectrometry
4.
J Med Food ; 23(10): 1019-1032, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32856978

ABSTRACT

The present study aimed to investigate the protective role of the flavonoid fisetin (FI) on inflammation-mediated metabolic diseases, especially tissue fibrosis and insulin resistance (IR) in high-fat diet (HFD)-induced obese mice. C57BL/6J mice were fed with normal-fat diet, HFD (40 kcal% fat), or HFD +0.02% (w/w) FI for 16 weeks. Dietary FI supplementation improved hepatic steatosis by restricting lipogenesis, while promoting lipolysis in the liver. FI also prevented adiposity via an increase in the expression of genes involved in FA oxidation and a decrease in the expression of genes involved in lipogenesis in white adipose tissue. In addition, FI increased brown adipose tissue (BAT) and skeletal muscle weights, thermogenic gene mRNA expression in BAT, and tricarboxylic acid cycle-related gene expression in skeletal muscle, which may be linked to the prevention of nonalcoholic fatty liver disease as well as adiposity. Moreover, FI supplementation decreased excessive reactive oxygen species production by increasing paraoxonase activity, adipokine dysregulation, proinflammatory cytokine production, and extracellular matrix amassment in the liver. FI supplementation ameliorated IR, in part, by normalizing pancreatic islet dysfunction, and it declined hepatic gluconeogenesis and proinflammatory responses. Taken together, the present findings indicate that FI can protect against HFD-induced inflammation-mediated disorders, including fibrosis and IR.


Subject(s)
Adipocytes/drug effects , Flavonols/pharmacology , Insulin Resistance , Liver Cirrhosis/drug therapy , Adipocytes/pathology , Adiposity/drug effects , Animals , Diet, High-Fat/adverse effects , Mice , Mice, Inbred C57BL , Mice, Obese
5.
Molecules ; 25(16)2020 Aug 11.
Article in English | MEDLINE | ID: mdl-32796637

ABSTRACT

d-allulose is an uncommon sugar that provides almost no calories when consumed. Its sweetness is 70% that of sucrose. d-allulose is a metabolic regulator of glucose and lipid metabolism. However, few reports concerning its effect on diabetes and related metabolic disturbances in db/db mice are available. In this study, we evaluated d-allulose's effect on hyperglycemia, hyperinsulinemia, diabetes and inflammatory responses in C57BL/KsJ-db/db mice. Mice were divided into normal diet, erythritol supplemented (5% w/w), and d-allulose supplemented (5% w/w) groups. Blood glucose and plasma glucagon levels and homeostatic model assessment (HOMA-IR) were significantly lower in the d-allulose group than in the normal diet group, and plasma insulin level was significantly increased. Further, d-allulose supplement significantly increased hepatic glucokinase activity and decreased hepatic phosphoenolpyruvate carboxykinase and glucose-6-phosphatase activity. Expression of glucose transporter 4, insulin receptor substrate 1, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha and AKT serine/threonine kinase 2 were also upregulated by d-allulose supplement in adipocyte and muscle. Finally, d-allulose effectively lowered plasma and hepatic triglyceride and free fatty acid levels, and simultaneously reduced hepatic fatty acid oxidation and carnitine palmitoyl transferase activity. These changes are likely attributable to suppression of hepatic fatty acid synthase and glucose-6-phosphate dehydrogenase activity. Notably, d-allulose also reduced pro-inflammatory adipokine and cytokine levels in plasma. Our results indicate that d-allulose is an effective sugar substitute for improving lipid and glucose metabolism.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Fructose/pharmacology , Hyperglycemia/drug therapy , Hyperinsulinism/drug therapy , Inflammation/drug therapy , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Hyperglycemia/metabolism , Hyperglycemia/pathology , Hyperinsulinism/metabolism , Hyperinsulinism/pathology , Inflammation/metabolism , Inflammation/pathology , Insulin/blood , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains
6.
Nutrients ; 12(7)2020 Jul 16.
Article in English | MEDLINE | ID: mdl-32708827

ABSTRACT

Allulose has been reported to serve as an anti-obesity and anti-diabetic food component; however, its molecular mechanism is not yet completely understood. This study aims to elucidate the mechanisms of action for allulose in obesity-induced type 2 diabetes mellitus (T2DM), by analyzing the transcriptional and microbial populations of diet-induced obese mice. Thirty-six C57BL/6J mice were divided into four groups, fed with a normal diet (ND), a high-fat diet (HFD), a HFD supplemented with 5% erythritol, or a HFD supplemented with 5% allulose for 16 weeks, in a pair-fed manner. The allulose supplement reduced obesity and comorbidities, including inflammation and hepatic steatosis, and changed the microbial community in HFD-induced obese mice. Allulose attenuated obesity-mediated inflammation, by downregulating mRNA levels of inflammatory response components in the liver, leads to decreased plasma pro-inflammatory marker levels. Allulose suppressed glucose and lipid metabolism-regulating enzyme activities, ameliorating hepatic steatosis and improving dyslipidemia. Allulose improved fasting blood glucose (FBG), plasma glucose, homeostatic model assessment of insulin resistance (HOMA-IR), and the area under the curve (AUC) for the intraperitoneal glucose tolerance test (IPGTT), as well as hepatic lipid levels. Our findings suggested that allulose reduced HFD-induced obesity and improved T2DM by altering mRNA expression and the microbiome community.


Subject(s)
Diabetes Mellitus, Type 2/microbiology , Diabetes Mellitus, Type 2/therapy , Diet, High-Fat/adverse effects , Dietary Supplements , Fructose/administration & dosage , Gastrointestinal Microbiome , Hypoglycemic Agents , Obesity/microbiology , Obesity/therapy , Phytotherapy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Animals , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Fructose/pharmacology , Glucose/metabolism , Inflammation , Inflammation Mediators/metabolism , Insulin Resistance , Lipid Metabolism , Liver/metabolism , Male , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism
7.
J Autoimmun ; 111: 102436, 2020 07.
Article in English | MEDLINE | ID: mdl-32220507

ABSTRACT

Low grade, chronic inflammation is a critical risk factor for immunologic dysfunction including autoimmune diseases. However, the multiplicity of complex mechanisms and lack of relevant murine models limit our understanding of the precise role of chronic inflammation. To address these hurdles, we took advantage of multi-omics data and a unique murine model with a low but chronic expression of IFN-γ, generated by replacement of the AU-rich element (ARE) in the 3' UTR region of IFN-γ mRNA with random nucleotides. Herein, we demonstrate that low but differential expression of IFN-γ in mice by homozygous or heterozygous ARE replacement triggers distinctive gut microbial alterations, of which alteration is female-biased with autoimmune-associated microbiota. Metabolomics data indicates that gut microbiota-dependent metabolites have more robust sex-differences than microbiome profiling, particularly those involved in fatty acid oxidation and nuclear receptor signaling. More importantly, homozygous ARE-Del mice have dramatic changes in tryptophan metabolism, bile acid and long-chain lipid metabolism, which interact with gut microbiota and nuclear receptor signaling similarly with sex-dependent metabolites. Consistent with these findings, nuclear receptor signaling, encompassing molecules such as PPARs, FXR, and LXRs, was detectable as a top canonical pathway in comparison of blood and tissue-specific gene expression between female homozygous vs heterozygous ARE-Del mice. Further analysis implies that dysregulated autophagy in macrophages is critical for breaking self-tolerance and gut homeostasis, while pathways interact with nuclear receptor signaling to regulate inflammatory responses. Overall, pathway-based integration of multi-omics data provides systemic and cellular insights about how chronic inflammation driven by IFN-γ results in the development of autoimmune diseases with specific etiopathological features.


Subject(s)
Autoimmune Diseases/immunology , Dysbiosis/immunology , Inflammation/immunology , Interferon-gamma/metabolism , Macrophages/immunology , 3' Untranslated Regions/genetics , AU Rich Elements/genetics , Animals , Autophagy , Chronic Disease , Female , Gastrointestinal Microbiome/immunology , Interferon-gamma/genetics , Male , Mice , Mice, Knockout , Receptors, Cytoplasmic and Nuclear/metabolism , Sexism , Signal Transduction
8.
J Med Food ; 23(3): 233-241, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32191577

ABSTRACT

Eriocitrin (EC) is an abundant flavonoid in lemons, which is known as a strong antioxidant agent. This study investigated the biological and molecular mechanisms underlying the anti-obesity effect of EC in high-fat diet (HFD)-fed obese mice. C57BL/6N mice were fed an HFD (40 kcal% fat) with or without 0.005% (w/w) EC for 16 weeks. Dietary EC improved adiposity by increasing adipocyte fatty acid (FA) oxidation, energy expenditure, and mRNA expression of thermogenesis-related genes in brown adipose tissue (BAT) and skeletal muscle, whereas it also decreased lipogenesis-related gene expression in white adipose tissue. In addition to adiposity, EC prevented hepatic steatosis by diminishing lipogenesis while enhancing FA oxidation in the liver and fecal lipid excretion, which was linked to attenuation of hyperlipidemia. Moreover, EC improved insulin sensitivity by decreasing hepatic gluconeogenesis and proinflammatory responses. These findings indicate that EC may protect against diet-induced adiposity and related metabolic disorders by controlling thermogenesis of BAT and skeletal muscle, FA oxidation, lipogenesis, fecal lipid excretion, glucose utilization, and gluconeogenesis.


Subject(s)
Adiposity/drug effects , Flavanones/administration & dosage , Obesity/drug therapy , Plant Extracts/administration & dosage , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/physiopathology , Animals , Citrus/chemistry , Diet, High-Fat , Lipogenesis/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Obesity/etiology , Obesity/metabolism , Obesity/physiopathology , Phytotherapy , Thermogenesis/drug effects
9.
Nutrients ; 12(2)2020 Jan 29.
Article in English | MEDLINE | ID: mdl-32013116

ABSTRACT

Recently, there has been a global shift in diet towards an increased intake of energy-dense foods that are high in sugars. D-allulose has received attention as a sugar substitute and has been reported as one of the anti-obesity food components; however, its correlation with the intestinal microbial community is not yet completely understood. Thirty-six C57BL/6J mice were divided in to four dietary groups and fed a normal diet (ND), a high-fat diet (HFD, 20% fat, 1% cholesterol, w/w), and a HFD with 5% erythritol (ERY) and D-allulose (ALL) supplement for 16 weeks. A pair-feeding approach was used so that all groups receiving the high-fat diet would have the same calorie intake. As a result, body weight and body fat mass in the ALL group were significantly decreased toward the level of the normal group with a simultaneous decrease in plasma leptin and resistin. Fecal short-chain fatty acid (SCFA) production analysis revealed that ALL induced elevated total SCFA production compared to the other groups. Also, ALL supplement induced the change in the microbial community that could be responsible for improving the obesity based on 16S rRNA gene sequence analysis, and ALL significantly increased the energy expenditure in Day(6a.m to 6pm). Taken together, our findings suggest that 5% dietary ALL led to an improvement in HFD-induced obesity by altering the microbiome community.


Subject(s)
Diet, High-Fat/adverse effects , Fructose/administration & dosage , Fructose/pharmacology , Gastrointestinal Microbiome/drug effects , Obesity/chemically induced , Obesity/drug therapy , Animals , Dietary Supplements , Male , Mice , Mice, Inbred C57BL
10.
Mol Nutr Food Res ; 64(5): e1900982, 2020 03.
Article in English | MEDLINE | ID: mdl-31845524

ABSTRACT

SCOPE: The results of recent studies on d-allulose intervention in high-fat diet (HFD)-fed mice suggest that d-allulose has a substantial impact on obesity. In addition, several studies have uncovered bacterial candidates among the gut microbiota associated with obesity and inflammation in mice. To identify the d-allulose-attenuated genes related to the inflammation-associated bacterial candidates, two types of statistical analyses are performed. METHODS AND RESULTS: Using liver and epididymal fat tissues, genes with expression levels that recovered from HFD-induced dysregulation are identified through differentially expressed gene (DEG) analysis. Finally, correlation-based network analysis between the diet, microbes, and the candidates identified from DEG analysis reveal 20 genes that showed anti-obesogenic patterns and associations with Lactobacillus and Coprococcus, which are representative bacterial candidates associated with inflammation and obesity. CONCLUSION: The results of the present study suggest that d-allulose closely interacts with the candidate genes and microbes to alleviate weight gain and inflammation, partly via down regulation of Gm12250 expression in multiple tissues and increases the Lactobacillus and Coprococcus in gut microbiota composition.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Fructose/pharmacology , Gastrointestinal Microbiome/drug effects , Gene Expression Regulation/drug effects , Obesity/microbiology , Animals , Anti-Obesity Agents/pharmacology , Diet, High-Fat , Dietary Supplements , Gastrointestinal Microbiome/physiology , Male , Mice, Inbred C57BL , Mice, Obese , Obesity/etiology , Obesity/genetics
11.
Nutrients ; 11(10)2019 Oct 14.
Article in English | MEDLINE | ID: mdl-31615016

ABSTRACT

Platycodon grandiflorus root extract (PGE) has shown various properties, such as anti-hyperlipidemia, anti-diabetic, and anti-obesity, but mostly in animal studies. Therefore, we conducted a preliminary study on the anti-obesity effect of PGE in 108 Korean adults (aged 20-60 years, 30 kg/m2 ≥ body mass index ≥ 23 kg/m2). The participants were randomly assigned to four groups and were administered the placebo, PGE571 (571 mg as PGE), PGE1142 (1142 mg as PGE), and PGE2855 (2855 mg as PGE), independently, for 12 weeks. Body composition, nutrient intake, computed tomography scan, and plasma adipokines, as well as hepatic/renal function markers, were assessed. The PGE571 group revealed a significant decrease in body fat mass and body fat percentage when compared with the placebo group. Moreover, the total abdominal and subcutaneous fat areas were significantly decreased following PGE (PGE2855 group) supplementation. These results provide useful information on the anti-obesity effect of PGE for overweight and obese adult humans.


Subject(s)
Overweight/drug therapy , Plant Extracts/pharmacology , Platycodon/chemistry , Adult , Double-Blind Method , Female , Humans , Male , Middle Aged , Plant Extracts/chemistry , Young Adult
12.
J Med Food ; 22(11): 1100-1109, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31566484

ABSTRACT

Adipocytes regulate lipid metabolism according to physiological energy requirements. A dysfunctional lipid metabolism can lead to obesity and its complications such as hepatic steatosis, diabetes, and hyperlipidemia. In our study, the impact of Platycodon grandiflorus root ethanol extract (PGH) on lipid excretion and thermogenesis-related markers in diet-induced obesity mice was analyzed. Our data show that PGH elevated fatty acid uptake in epididymal adipose tissue by increasing Cd36, Slc27a1, Ffar2, and Ffar4 expression, which led to decreased blood free fatty acid concentrations. Moreover, PGH normalized body weight and fat mass in diet-induced obese mice by increasing lipolysis (Plin1, Atgl, and Hsl) and fatty acid oxidation. Changes in the levels of browning-related genes, enzyme activity of carnitine palmitoyltransferase, and the overall transcriptome (Bmp4, Cidec, Ucp3, Sirt3, and Cox4i1) led to promote brown adipose tissue-like features (browning) in epididymal white adipose tissue and enhanced energy expenditure. Our results suggest that PGH promotes lipid excretion and thermogenic function in high-fat diet-induced obese mice, which are mediated by regulation of fat metabolism.


Subject(s)
Lipid Metabolism , Lipolysis , Plant Extracts/pharmacology , Platycodon/chemistry , Thermogenesis , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Body Weight , Energy Metabolism , Fatty Acids/metabolism , Gene Expression Regulation , Male , Mice, Inbred C57BL , Mice, Obese , Plant Roots/chemistry , Random Allocation , Transcriptome
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.
J Med Food ; 22(9): 928-936, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31390281

ABSTRACT

Bangpungtongseong-san (BT), an oriental herbal medicine, is used to treat obesity in Korea and East Asia and its antiobesity effects have been examined by several researchers. However, the molecular mechanisms of the antihepatic steatosis effects of BT are unclear. In this study, we examined the effects of BT on obesity, particularly nonalcoholic fatty liver disease, by analyzing metabolic and transcriptional responses using mRNA-sequencing profiles. C57BL/6J mice were fed a high-fat diet (HFD) or HFD + BT (1.5%, w/w, BT) for 12 weeks. Phenotype characteristics were estimated, and the antiobesity mechanism was examined using mRNA sequencing transcriptomic profiles in HFD-induced obese mice. BT treatment ameliorated dyslipidemia and hepatic steatosis in HFD-induced obese mice and reduced body weight gain. The levels of hepatic lipotoxicity markers were significantly decreased, while hepatic antioxidant enzyme activities were augmented by BT compared with in the HFD group. BT attenuated HFD-induced fatty liver through transcriptional changes in the liver. BT treatment downregulated mitochondrial oxidative phosphorylation-related genes in the liver, suggesting improved mitochondrial function. BT treatment also decreased the hepatic fibrosis-related transcriptome. Our findings provide insight into the antiobesity effects of BT, an alternative oriental medicine, for treating obesity-related conditions. Metabolic and transcriptional responses to diet-induced obesity with BT treatment improved liver function.


Subject(s)
Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/genetics , Obesity/drug therapy , Obesity/genetics , Plant Extracts/administration & dosage , Animals , Diet, High-Fat/adverse effects , Humans , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/etiology , Obesity/metabolism , Phytotherapy , Plants, Medicinal/chemistry , Transcription, Genetic/drug effects
15.
Nutrients ; 11(6)2019 Jun 15.
Article in English | MEDLINE | ID: mdl-31208033

ABSTRACT

This study aimed to elucidate the molecular mechanism of Chrysanthemum morifolium Ramat. against obesity and diabetes, by comparing the transcriptional changes in epididymal white adipose tissue (eWAT) with those of the bioactive compound in C. morifolium, luteolin (LU). Male C57BL/6J mice were fed a normal diet, high-fat diet (HFD), and HFD supplemented with 1.5% w/w chrysanthemum leaf ethanol extract (CLE) for 16 weeks. Supplementation with CLE and LU significantly decreased the body weight gain and eWAT weight by stimulating mRNA expressions for thermogenesis and energy expenditure in eWAT via lipid mobilization, which may be linked to the attenuation of dyslipidemia. Furthermore, CLE and LU increased uncoupling protein-1 protein expression in brown adipose tissue, leading to energy expenditure. Of note, CLE and LU supplements enhanced the balance between lipid storage and mobilization in white adipose tissue (WAT), in turn, inhibiting adipocyte inflammation and lipotoxicity of peripheral tissues. Moreover, CLE and LU attenuated hepatic steatosis by suppressing hepatic lipogenesis, thereby ameliorating insulin resistance and dyslipidemia. Our data suggest that CLE helps inhibit obesity and its comorbidities via the complex interplay between liver and WAT in diet-induced obese mice.


Subject(s)
Adipose Tissue, White/drug effects , Chrysanthemum/chemistry , Dietary Supplements , Ethanol/pharmacology , Lipid Mobilization/drug effects , Metabolic Diseases/prevention & control , Obesity/prevention & control , Phytotherapy , Adipose Tissue, Brown/metabolism , Animals , Diet, High-Fat , Energy Metabolism , Insulin Resistance , Liver/metabolism , Male , Metabolic Diseases/etiology , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/etiology , Plant Extracts/pharmacology , Plant Leaves/chemistry
16.
Nutr Metab (Lond) ; 16: 31, 2019.
Article in English | MEDLINE | ID: mdl-31123488

ABSTRACT

BACKGROUND: Obesity increases the risk of metabolic dysfunction such as dyslipidemia, hypertension, and fatty liver. Physcion (PY) is an anthraquinone that reportedly has anti-inflammatory and anti-bacterial properties. However, few studies have addressed the effect of PY on high-fat diet-induced obesity in mice. The purpose of this study was to investigate the effects of PY on obesity. METHODS: Male C57BL/6 J mice were randomly divided into three groups and fed normal diet (ND, 5% fat, w/w), high-fat diet (HFD, 20% fat, 1% cholesterol, w/w), and HFD supplemented with 0.002% PY (w/w) for 16 weeks. Obesity-related biomarkers were analyzed including whole body and white adipose tissue (WAT) weight, in addition to lipid and inflammatory factors in the plasma, feces, liver and epididymal WAT. Significant differences among the groups were determined using Student's t-test. Differences were considered statistically significant at p < 0.05. RESULTS: Body and WAT weights were significantly decreased by the PY supplement relative to the HFD groups. Energy expenditure was enhanced by the PY supplement, which led to ameliorate plasma lipids, adipokines, cytokines, and fecal lipids. Fatty acid (FA) synthesis decreased in the liver, while FA oxidation increased. Finally, lipid synthesis markedly decreased whereas lipolysis and oxidation increased in WAT. CONCLUSIONS: The PY supplement suppressed lipid accumulation in WAT and the liver by regulating enzyme and gene levels. These results indicate that PY can improve diet-induced obesity and its complications such as dyslipidemia, hepatic steatosis, and inflammation.

17.
Int J Mol Sci ; 20(5)2019 Mar 11.
Article in English | MEDLINE | ID: mdl-30862092

ABSTRACT

The present study aimed to investigate the molecular mechanisms underlying the anti-obesity effect of flavonoid eriodictyol (ED) supplementation in mice fed with a high-fat diet (HFD). C57BL/6N mice were fed with normal diet (ND), HFD (40 kcal% fat), or HFD + 0.005% (w/w) ED for 16 weeks. In HFD-induced obese mice, dietary ED supplementation significantly alleviated dyslipidemia and adiposity by downregulating the expression of lipogenesis-related genes in white adipose tissue (WAT), while enhancing fecal lipid excretion. ED additionally improved hepatic steatosis and decreased the production of pro-inflammatory cytokines by downregulating the expression of hepatic enzymes and the genes involved in lipogenesis and upregulating the expression of hepatic fatty acid oxidation-related enzymes and genes. In addition, ED improved insulin resistance (IR) by suppressing hepatic gluconeogenesis, enhancing glucose utilization, and modulating the production and release of two incretin hormones, namely gastric inhibitory polypeptide (GIP) and glucagon-like peptide-1 (GLP-1). Taken together, the current findings indicated that ED can protect against diet-induced obesity and related metabolic disturbances, including dyslipidemia, inflammation, fatty liver disease, and IR in diet-induced obese mice.


Subject(s)
Adiposity/drug effects , Dietary Supplements , Fatty Liver/metabolism , Flavanones/pharmacology , Inflammation/metabolism , Insulin Resistance , Obesity/etiology , Obesity/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Adipokines/metabolism , Animals , Blood Glucose/drug effects , Body Weight/drug effects , Cytokines/metabolism , Diet, High-Fat/adverse effects , Disease Notification , Fatty Liver/drug therapy , Fatty Liver/etiology , Gene Expression Regulation/drug effects , Inflammation/drug therapy , Inflammation/etiology , Lipid Metabolism/drug effects , Mice , Mice, Obese , Obesity/drug therapy
18.
Nutrients ; 10(12)2018 Dec 19.
Article in English | MEDLINE | ID: mdl-30572580

ABSTRACT

D-allulose has recently received attention as a sugar substitute. However, there are currently no reports regarding its association with gastrointestinal (GI) tolerance. Thus, we performed a GI tolerance test for D-allulose in order to establish its daily acceptable intake level. When the dose of D-allulose was gradually increased in steps of 0.1 g/kg·Body Weight (BW) to identify the maximum single dose for occasional ingestion, no cases of severe diarrhea or GI symptoms were noted until a dose of 0.4 g/kg·BW was reached. Severe symptoms of diarrhea were noted at a dose of 0.5 g/kg·BW. Similarly, the GI tolerance test did not show any incidences of severe diarrhea or GI symptoms until a dose of 0.5 g/kg·BW was reached. A correlation analysis of the GI tolerance test for D-allulose and sugar revealed significantly higher frequencies of symptoms of diarrhea (p = 0.004), abdominal distention (p = 0.039), and abdominal pain (p = 0.031) after D-allulose intake. Increasing the total daily D-allulose intake gradually to 1.0 g/kg·BW for regular ingestion resulted in incidences of severe nausea, abdominal pain, headache, anorexia, and diarrheal symptoms. Based on these results, we suggest a maximum single dose and maximum total daily intake of D-Allulose of 0.4 g/kg·BW and 0.9 g/kg·BW, respectively.


Subject(s)
Abdominal Pain/chemically induced , Diarrhea/chemically induced , Fructose/adverse effects , Sweetening Agents/adverse effects , Adult , Dose-Response Relationship, Drug , Female , Fructose/administration & dosage , Humans , Male , Nausea/chemically induced , Sweetening Agents/administration & dosage , Young Adult
19.
Nutrients ; 10(11)2018 Nov 19.
Article in English | MEDLINE | ID: mdl-30463250

ABSTRACT

The effects of allulose and two probiotic species on diet-induced obese (DIO) mice were investigated. Lactobacillus sakei LS03 (108 cfu/day) and Leuconostoc kimchii GJ2 (108 cfu/day) were used as probiotics, and allulose (AL) as a prebiotic. The synergistic effect of prebiotics and probiotics in improving obesity was evaluated. Orally fed Lactobacillus sakei LS03 (LS) or Leuconostoc kimchii GJ2 (GJ), significantly decreased hepatic triglyceride (TG) and fatty acid (FA) compared to the high-fat diet (HFD) control. AL markedly decreased visceral adiposity and pro-inflammatory adipokines (leptin and resistin) and cytokines (IL-6 and IL-1ß) as well as hepatic TG and FA. In addition, AL exerted synergic effects with probiotics (LS and/or GJ) on the reduction of visceral white adipose tissue (WAT), associated with a decreased leptin: adiponectin ratio. There was no significant differences between the AL-SL and AL group, allulose and GJ combination (AL-GJ) was more effective than allulose in improving dyslipidemia, and decreasing WAT weight and hepatic FA, suggesting allulose may act as a favorable prebiotic for GJ supplement than LS. Combination of allulose with LS and GJ supplementation (AL-LSGJ) was the most effective for improving obesity related complications among the synbiotics groups containing allulose. In conclusion, this study demonstrated that the synbiotic mixture with allulose was more effective in suppressing diet-induced obese (DIO) and its complications via the regulation of lipid metabolism, than the probiotics or allulose alone, suggesting allulose may act as a prebiotic for the two probiotics tested in the study. This new synbiotic mixture with allulose may help ameliorate the deleterious effects of diet-induced obesity and contribute to the growth of the food industry.


Subject(s)
Adiposity , Fructose/analysis , Synbiotics , Animals , Biomarkers/blood , Cytokines/blood , Diet, High-Fat , Latilactobacillus sakei , Leptin/metabolism , Leuconostoc , Lipid Metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/therapy , Prebiotics , Probiotics , Triglycerides/blood , Weight Loss
20.
Nutrients ; 10(11)2018 Nov 09.
Article in English | MEDLINE | ID: mdl-30423963

ABSTRACT

The aim of the current study was to elucidate the effects of long-term supplementation with dietary ursolic acid (UR) on obesity and associated comorbidities by analyzing transcriptional and metabolic responses, focusing on the role of UR in the modulation of the circadian rhythm pathway in particular. C57BL/6J mice were divided into three groups and fed a normal diet, high-fat diet, or high-fat + 0.05% (w/w) UR diet for 16 weeks. Oligonucleotide microarray profiling revealed that UR is an effective regulator of the liver transcriptome, and canonical pathways associated with the "circadian rhythm" and "extracellular matrix (ECM)⁻receptor interactions" were effectively regulated by UR in the liver. UR altered the expression of various clock and clock-controlled genes (CCGs), which may be linked to the improvement of hepatic steatosis and fibrosis via lipid metabolism control and detoxification enhancement. UR reduced excessive reactive oxygen species production, adipokine/cytokine dysregulation, and ECM accumulation in the liver, which also contributed to improve hepatic lipotoxicity and fibrosis. Moreover, UR improved pancreatic islet dysfunction, and suppressed hepatic gluconeogenesis, thereby reducing obesity-associated insulin resistance. Therapeutic approaches targeting hepatic circadian clock and CCGs using UR may ameliorate the deleterious effects of diet-induced obesity and associated complications such as hepatic fibrosis.


Subject(s)
Circadian Rhythm/drug effects , Diet, High-Fat/adverse effects , Fatty Liver/drug therapy , Insulin Resistance , Liver Cirrhosis/drug therapy , Obesity/drug therapy , Triterpenes/therapeutic use , Adipokines/metabolism , Animals , Circadian Clocks , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/therapeutic use , Cytokines/metabolism , Fatty Liver/metabolism , Gluconeogenesis/drug effects , Islets of Langerhans/drug effects , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Male , Mice, Inbred C57BL , Mice, Obese , Obesity/etiology , Obesity/metabolism , Phytotherapy , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Reactive Oxygen Species/metabolism , Receptors, Cell Surface/metabolism , Triterpenes/metabolism , Triterpenes/pharmacology , Ursolic Acid
SELECTION OF CITATIONS
SEARCH DETAIL