Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Life Sci ; 298: 120488, 2022 Jun 01.
Article in English | MEDLINE | ID: mdl-35331721

ABSTRACT

AIM: Increasing brown adipocytes activity and inducting browning of white adipocytes are potential therapeutic targets for the treatment of obesity. In the present study, we investigated the effects of Tanshinone 1 (Tan 1), a major compound from Salvia miltiorrhiza Bunge, on the activation of brown adipocytes and browning of white adipocytes in vivo and in vitro. MATERIALS AND METHODS: Expression of genes associated with brown adipocyte function including thermogenesis, mitochondria biogenesis and fatty acid oxidation was examined in brown adipose tissue (BAT) and white adipose tissue (WAT) of high fat diet (HFD)-fed obese mice administrated with Tan 1 or in immortalized brown adipocytes (iBAs) and 3T3-L1 adipocytes treated with Tan 1. Mitochondria DNA (mtDNA) content, lipolysis and phosphorylated AMP-activated protein kinase (AMPK) were further assessed in Tan 1 treated-iBAs and 3T3-L1 adipocytes. KEY FINDINGS: The administration of Tan 1 protected against HFD-induced obesity in mice, which was associated with enhanced expression of brown adipocyte function-related genes in BAT and WAT. Tan 1 treatment also upregulated brown adipocyte function-related genes in iBA and induced beige adipocytes genes in 3T3-L1 adipocytes, resulting in increased mtDNA content and lipolysis. Tan 1 activated AMPK in BAT and WAT of HFD-fed obese mice as well as in iBAs and 3T3-L1 adipocytes. Inhibition of AMPK by compound C prevented Tan 1-induced expression of beige adipocytes genes. SIGNIFICANCE: These results indicate that Tan 1 activates brown adipocytes and induces browning of white adipocytes, which may contribute to anti-obesity activity of Tan 1.


Subject(s)
Abietanes , Adipocytes, Brown , Diet, High-Fat , Obesity , Animals , Mice , 3T3-L1 Cells , Abietanes/pharmacology , Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , AMP-Activated Protein Kinases/metabolism , Diet, High-Fat/adverse effects , DNA, Mitochondrial/metabolism , Mice, Obese , Obesity/drug therapy , Obesity/etiology , Obesity/prevention & control , Thermogenesis
2.
Int J Mol Sci ; 22(20)2021 Oct 14.
Article in English | MEDLINE | ID: mdl-34681759

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease. While the development of NAFLD is correlated with aberrant histone methylation, modifiers of histone methylation involved in this event remain poorly understood. Here, we studied the functional role of the histone demethylase KDM7A in the development of hepatic steatosis. KDM7A overexpression in AML12 cells upregulated diacylglycerol acyltransferase 2 (DGAT2) expression and resulted in increased intracellular triglyceride (TG) accumulation. Conversely, KDM7A knockdown reduced DGAT2 expression and TG accumulation, and significantly reversed free fatty acids-induced TG accumulation. Additionally, adenovirus-mediated overexpression of KDM7A in mice resulted in hepatic steatosis, which was accompanied by increased expression of hepatic DGAT2. Furthermore, KDM7A overexpression decreased the enrichment of di-methylation of histone H3 lysine 9 (H3K9me2) and H3 lysine 27 (H3K27me2) on the promoter of DGAT2. Taken together, these results indicate that KDM7A overexpression induces hepatic steatosis through upregulation of DGAT2 by erasing H3K9me2 and H3K27me2 on the promoter.


Subject(s)
Diacylglycerol O-Acyltransferase/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Animals , Diacylglycerol O-Acyltransferase/genetics , Hep G2 Cells , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Lysine/metabolism , Mice, Inbred C57BL , Mice, Obese , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Promoter Regions, Genetic , Triglycerides/metabolism
3.
Int J Mol Sci ; 21(21)2020 Nov 05.
Article in English | MEDLINE | ID: mdl-33167594

ABSTRACT

Ligand-activated liver X receptor α (LXRα) upregulates the expression of hepatic lipogenic genes, which leads to triglyceride (TG) accumulation, resulting in nonalcoholic fatty liver disease (NAFLD). Thus, LXRα regulation may provide a novel therapeutic target against NAFLD. However, histone methylation-mediated epigenetic regulation involved in LXRα-dependent lipogenesis is poorly understood. In this study, we investigated the functional role of the histone demethylase Jumonji domain-containing protein 2B (JMJD2B) in LXRα-dependent lipogenesis. JMJD2B expression level was upregulated in HepG2 cells treated with LXRα agonist T0901317 or palmitate and the liver of mice administered with T0901317 or fed a high-fat diet. Knockdown of JMJD2B using siRNA abrogated T0901317-induced LXRα-dependent lipogenic gene expression and lowered intracellular TG accumulation. Conversely, overexpression of JMJD2B in HepG2 cells upregulated the expression of LXRα-dependent lipogenic genes, in line with increased intracellular TG levels. JMJD2B overexpression or T0901317 treatment induced the recruitment of JMJD2B and LXRα to LXR response elements (LXRE) in the promoter region of LXRα-target gene and reduced the enrichment of H3K9me2 and H3K9me3 in the vicinity of the LXRE. Furthermore, JMJD2B enhanced T0901317 or LXRα-induced transcriptional activities of reporters containing LXRE. A co-immunoprecipitation assay revealed that JMJD2B interacted with activated LXRα. Moreover, overexpression of JMJD2B in mice resulted in upregulation of hepatic LXRα-dependent lipogenic genes, consistent with development of hepatic steatosis. Taken together, these results indicate that JMJD2B plays a role in LXRα-mediated lipogenesis via removing the repressive histone marks, H3K9me2 and H3K9me3, at LXRE, which might contribute to hepatic steatosis.


Subject(s)
Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Lipogenesis/physiology , Liver X Receptors/metabolism , Animals , Diet, High-Fat , Epigenesis, Genetic , Female , Hep G2 Cells , Hepatocytes/metabolism , Histone Demethylases/genetics , Histones/metabolism , Humans , Hydrocarbons, Fluorinated/pharmacology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Palmitates/pharmacology , Sterol Regulatory Element Binding Protein 1/metabolism , Sulfonamides/pharmacology , Transcriptional Activation
4.
Nutr Res Pract ; 14(4): 423, 2020 08.
Article in English | MEDLINE | ID: mdl-32765821

ABSTRACT

[This retracts the article on p. 15 in vol. 7, PMID: 23423690.].

5.
Nutrients ; 12(5)2020 Apr 27.
Article in English | MEDLINE | ID: mdl-32349456

ABSTRACT

Tanshinone I (Tan I) is a diterpenoid isolated from Salvia miltiorrhiza Bunge and exhibits antitumor effects in several cancers. However, the anti-obesity properties of Tan I remain unexplored. Here, we evaluated the anti-obesity effects of Tan I in high-fat-diet (HFD)-induced obese mice and investigated the underlying molecular mechanisms in 3T3-L1 cells. HFD-induced obese mice were orally administrated Tan I for eight weeks, and body weight, weight gain, hematoxylin and eosin staining and serum biological parameters were examined. The adipogenesis of 3T3-L1 preadipocytes was assessed using Oil Red O staining and measurement of intracellular triglyceride (TG) levels, and mitotic clonal expansion (MCE) and its related signal molecules were analyzed during early adipogenesis of 3T3-L1 cells. The administration of Tan I significantly reduced body weight, weight gain, and white adipocyte size, and improved obesity-induced serum levels of glucose, free fatty acid, total TG, and total cholesterol in vivo in HFD-induced obese mice. Furthermore, Tan I-administered mice demonstrated improvement of glucose metabolism and insulin sensitivity. Treatment with Tan I inhibited the adipogenesis of 3T3-L1 preadipocytes in vitro, with this inhibition mainly occurring at an early phase of adipogenesis through the attenuation of MCE via cell cycle arrest at the G1/S phase transition. Tan I inhibited the phosphorylation of p38, extracellular signal-regulated kinase (ERK), and Akt during the process of MCE, while it stimulated the phosphorylation of AMP-activated protein kinase. Furthermore, Tan I repressed the expression of CCAAT-enhancer-binding protein ß (C/EBPß), histone H3K9 demethylase JMJD2B, and subsequently cell cycle genes. Moreover, Tan I regulated the expression of early adipogenic transcription factors including GATAs and Kruppel-like factor family factors. These results indicate that Tan I prevents HFD-induced obesity via the inhibition of early adipogenesis, and thus improves glucose metabolism and insulin sensitivity. This suggests that Tan I possesses therapeutic potential for the treatment of obesity and obesity-related diseases.


Subject(s)
Abietanes/pharmacology , Adipogenesis/drug effects , Anti-Obesity Agents , Diet, High-Fat/adverse effects , Obesity/drug therapy , Phytotherapy , Salvia miltiorrhiza/chemistry , 3T3 Cells , AMP-Activated Protein Kinases/metabolism , Abietanes/administration & dosage , Abietanes/isolation & purification , Adipocytes/metabolism , Adipocytes/physiology , Adipogenesis/genetics , Administration, Oral , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Cycle/genetics , Depression, Chemical , Glucose/metabolism , Mice , Obesity/etiology , Obesity/metabolism , Phosphorylation , Triglycerides/metabolism
6.
Int J Mol Sci ; 20(19)2019 Sep 27.
Article in English | MEDLINE | ID: mdl-31569635

ABSTRACT

Poria cocos Wolf (PCW) is an edible, pharmaceutical mushroom with remarkable biological properties including anti-tumor, anti-inflammation, anti-oxidation, anti-ageing, and anti-diabetic effects. In the current study, we investigated the effects of PCW extract on hepatic steatosis under in vitro and in vivo conditions, and elucidated the underlying mechanisms. In this study, a mixture of HepG2 cells treated with free fatty acid (FFA)-palmitic and oleic acid-and high-fat diet (HFD)-fed obese mice were used; in this background, the triglyceride (TG) levels in HepG2 cells and mice liver were measured, and the expression levels of genes associated with lipogenesis, fatty acid oxidation, endoplasmic reticulum (ER) stress, and autophagy were determined. Treatment of HepG2 cells with FFA enhanced intracellular TG levels in HepG2 cells, but co-treatment with PCW significantly attenuated the TG levels. Notably, PCW significantly enhanced the phosphorylation of AMP-activated protein kinase (AMPK), acetyl-CoA carboxylase (ACC), and sterol regulatory element-binding protein-1c (SREBP-1c) in FFA-treated HepG2 cells. PCW downregulated the expression of lipogenesis-related genes, but upregulated the expression of genes associated with fatty acid oxidation. Further, PCW inhibited FFA-induced expression of ER stress markers and induced autophagy proteins. However, inhibition of AMPK significantly attenuated the beneficial effects of PCW in HepG2 cells. Moreover, PCW efficiently decreased HFD-induced hepatic TG accumulation in vivo and increased the phosphorylation of hepatic AMPK. Three compounds present in PCW including poricoic acid, pachymic acid, and ergosterol, significantly decreased FFA-induced increase in intracellular TG levels, consistent with increased AMPK phosphorylation, suggesting that poricoic acid, pachymic acid, and ergosterol are responsible for PCW-mediated amelioration of hepatic steatosis. Taken together, these results demonstrated that PCW ameliorates hepatic steatosis through the regulation of lipid metabolism, inhibition of ER stress, and activation of autophagy in an AMPK-dependent manner. This suggested that PCW can be potentially used for the treatment of hepatic steatosis.


Subject(s)
Agaricales/chemistry , Autophagy/drug effects , Cell Extracts/pharmacology , Endoplasmic Reticulum Stress/drug effects , Lipid Metabolism/drug effects , MAP Kinase Signaling System/drug effects , Poria/chemistry , Animals , Cell Extracts/chemistry , Cell Survival/drug effects , Disease Models, Animal , Fatty Acids/metabolism , Fatty Acids/pharmacology , Fatty Liver/etiology , Fatty Liver/metabolism , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Lipogenesis/drug effects , Lipogenesis/genetics , Male , Mice , Protein Kinase Inhibitors/pharmacology
7.
Int J Mol Sci ; 20(9)2019 Apr 29.
Article in English | MEDLINE | ID: mdl-31035653

ABSTRACT

Endogenous cannabinoids (ECs) are lipid-signaling molecules that specifically bind to cannabinoid receptor types 1 and 2 (CB1R and CB2R) and are highly expressed in central and many peripheral tissues under pathological conditions. Activation of hepatic CB1R is associated with obesity, insulin resistance, and impaired metabolic function, owing to increased energy intake and storage, impaired glucose and lipid metabolism, and enhanced oxidative stress and inflammatory responses. Additionally, blocking peripheral CB1R improves insulin sensitivity and glucose metabolism and also reduces hepatic steatosis and body weight in obese mice. Thus, targeting EC receptors, especially CB1R, may provide a potential therapeutic strategy against obesity and insulin resistance. There are many CB1R antagonists, including inverse agonists and natural compounds that target CB1R and can reduce body weight, adiposity, and hepatic steatosis, and those that improve insulin sensitivity and reverse leptin resistance. Recently, the use of CB1R antagonists was suspended due to adverse central effects, and this caused a major setback in the development of CB1R antagonists. Recent studies, however, have focused on development of antagonists lacking adverse effects. In this review, we detail the important role of CB1R in hepatic insulin resistance and the possible underlying mechanisms, and the therapeutic potential of CB1R targeting is also discussed.


Subject(s)
Insulin Resistance , Receptor, Cannabinoid, CB1/metabolism , Animals , Disease Susceptibility , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Lipid Metabolism , Molecular Targeted Therapy , Muscle, Skeletal/metabolism , Obesity/etiology , Obesity/metabolism , Organ Specificity , Signal Transduction
8.
Int J Mol Sci ; 21(1)2019 Dec 30.
Article in English | MEDLINE | ID: mdl-31906014

ABSTRACT

Cryptotanshinone (CT), a diterpene that is isolated from Salvia miltiorrhiza Bunge, exhibits anti-cancer, anti-oxidative, anti-fibrosis, and anti-inflammatory properties. Here, we examined whether CT administration possess a hepatoprotective effect on chronic ethanol-induced liver injury. We established a chronic alcohol feeding mouse model while using C57BL/6 mice, and examined the liver sections with hematoxylin-eosin (H&E) and Oil Red O (ORO) staining. Further, we analyzed the lipogenesis, fatty acid oxidation, oxidative stress, and inflammation genes by using quantitative polymerase chain reaction (qPCR) and immunoblotting in in vivo, and in vitro while using HepG2 and AML-12 cells. CT treatment significantly ameliorated ethanol-promoted hepatic steatosis, which was consistent with the decreased hepatic triglyceride levels. Interestingly, CT activated the phosphorylation of AMP-activated protein kinase (AMPK), sirtuin 1 (SIRT1), and nuclear factor E2-related factor 2 (Nrf2) proteins. Importantly, compound C (AMPK inhibitor) significantly blocked the CT-mediated reduction in TG accumulation, but not Ex52735 (SIRT1 inhibitor), which suggested that CT countering ethanol-promoted hepatic steatosis is mediated by AMPK activation. Furthermore, CT significantly inhibited cytochrome P450 2E1 (CYP2E1) and enhanced both the expression of antioxidant genes and hepatic glutathione levels. Finally, CT inhibited the ethanol-induced inflammation in ethanol-fed mice and HepG2 cells. Overall, CT exhibits a hepatoprotective effect against ethanol-induced liver injury by the inhibition of lipogenesis, oxidative stress, and inflammation through the activation of AMPK/SIRT1 and Nrf2 and the inhibition of CYP2E1. Therefore, CT could be an effective therapeutic agent for treating ethanol-induced liver injury.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Chemical and Drug Induced Liver Injury, Chronic/drug therapy , Ethanol/adverse effects , NF-E2-Related Factor 2/metabolism , Phenanthrenes/pharmacology , Salvia miltiorrhiza/chemistry , Signal Transduction/drug effects , Sirtuin 1/metabolism , Animals , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Chemical and Drug Induced Liver Injury, Chronic/pathology , Disease Models, Animal , Fatty Liver , Glutathione/metabolism , Hep G2 Cells , Humans , Inflammation/genetics , Lipid Metabolism/genetics , Lipogenesis/genetics , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Phenanthrenes/therapeutic use
9.
Sci Rep ; 8(1): 13734, 2018 09 13.
Article in English | MEDLINE | ID: mdl-30214048

ABSTRACT

Understanding the epigenetic mechanisms underlying the progression of hepatic steatosis is important for identifying new therapeutic targets against nonalcoholic fatty liver disease (NAFLD). We investigated the functional role of histone demethylase JMJD2B in the pathologic regulation of hepatic steatosis. JMJD2B expression was markedly increased in HepG2 cells treated with palmitate and oleate or liver X receptor agonist T09013178 and in the liver of high-fat diet (HFD)-induced obese mice. Overexpression of JMJD2B using adenovirus in HepG2 cells stimulated the expression of peroxisome proliferator-activated receptor γ2 (PPARγ2) and its steatosis target genes associated with fatty acid uptake and lipid droplet formation, resulting in increased intracellular triglyceride (TG) accumulation. Conversely, knocking down JMJD2B using siRNA reversed JMJD2B-mediated effects in HepG2 cells. The JMJD2B-dependent upregulation of PPARγ2 was associated with the removal of di- and trimethylation of histone H3 lysine 9 on the promoter of PPARγ2. Furthermore, exogeneous expression of JMJD2B using adenovirus in mice resulted in hepatic steatosis when fed a HFD, which was accompanied with increased expression of hepatic PPARγ2 and its steatosis target genes. Together, our results provide novel insights into the pivotal role of JMJD2B in the development of hepatic steatosis through upregulation of PPARγ2 and steatosis target genes.


Subject(s)
Fatty Liver/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Non-alcoholic Fatty Liver Disease/genetics , PPAR gamma/genetics , Animals , Diet, High-Fat , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Expression Regulation , Hep G2 Cells , Humans , Liver/enzymology , Liver/metabolism , Mice , Mice, Obese , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Oleic Acid/metabolism , Palmitic Acids/metabolism , Triglycerides/genetics
10.
Oncol Lett ; 16(4): 4193-4200, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30197667

ABSTRACT

The stembark of Sorbus commixta Hedl. has been used for treating asthma, bronchitis, gastritis and edema. However, the anticancer and proapoptotic effects of the water extract of the stembark of S. commixta (SCE) remain unknown. In the present study, it was shown that SCE inhibited the cell viability of the hepatocellular carcinoma cell lines Hep3B and HepG2, and of the colon carcinoma cell line HCT116. DNA content analysis indicated that SCE increased the sub-G1 population of HCT116 cells. In addition, degradation of nuclear DNA and levels of proapoptotic cascade components, including caspase-9, caspase-3 and poly ADP-ribose polymerase, were augmented by SCE treatment. Mitochondrial membrane potential and the ratio of B-cell lymphoma-2 (Bcl-2)/Bcl-2-associated X protein (Bax) were also reduced. Furthermore, SCE increased the expression of proapoptotic proteins, including p21, p27 and p53. Mouse double minute 2 homology, a negative regulator of p53, was cleaved by SCE treatment. Intracellular reactive oxygen species (ROS) production was also increased by SCE treatment. However, the SCE-induced cytotoxic effects and the increased expression of proapoptotic proteins, including p53 and p21, and reduced Bcl-2/Bax ratio, could be attenuated by N-acetyl cysteine, an ROS inhibitor. Taken together, these results indicate that SCE is a potent proapoptotic herbal medicine, which exerts its effects via the ROS-mediated mitochondrial pathway.

11.
Int J Mol Sci ; 19(9)2018 Sep 01.
Article in English | MEDLINE | ID: mdl-30200508

ABSTRACT

Gomisin N (GN), a lignan derived from Schisandra chinensis, has been shown to possess antioxidant, anti-inflammatory, and anticancer properties. In the present study, we investigated the protective effect of GN against ethanol-induced liver injury using in vivo and in vitro experiments. Histopathological examination revealed that GN administration to chronic-binge ethanol exposure mice significantly reduced ethanol-induced hepatic steatosis through reducing lipogenesis gene expression and increasing fatty acid oxidation gene expression, and prevented liver injury by lowering the serum levels of aspartate transaminase and alanine transaminase. Further, it significantly inhibited cytochrome P450 2E1 (CYP2E1) gene expression and enzyme activity, and enhanced antioxidant genes and glutathione level in hepatic tissues, which led to decreased hepatic malondialdehyde levels. It also lowered inflammation gene expression. Finally, GN administration promoted hepatic sirtuin1 (SIRT1)-AMP-activated protein kinase (AMPK) signaling in ethanol-fed mice. Consistent with in vivo data, treatment with GN decreased lipogenesis gene expression and increased fatty acid oxidation gene expression in ethanol-treated HepG2 cells, thereby preventing ethanol-induced triglyceride accumulation. Furthermore, it inhibited reactive oxygen species generation by downregulating CYP2E1 and upregulating antioxidant gene expression, and suppressed inflammatory gene expression. Moreover, GN prevented ethanol-mediated reduction in SIRT1 and phosphorylated AMPK. These findings indicate that GN has therapeutic potential against alcoholic liver disease through inhibiting hepatic steatosis, oxidative stress and inflammation.


Subject(s)
Fatty Liver, Alcoholic/metabolism , Lignans/pharmacology , Lipogenesis/drug effects , Liver/drug effects , Oxidative Stress/drug effects , Polycyclic Compounds/pharmacology , Alanine Transaminase/blood , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Aspartate Aminotransferases/blood , Cyclooctanes/administration & dosage , Cyclooctanes/pharmacology , Ethanol/toxicity , Fatty Liver, Alcoholic/drug therapy , Hep G2 Cells , Humans , Lignans/administration & dosage , Liver/injuries , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Polycyclic Compounds/administration & dosage
12.
Int J Mol Sci ; 19(4)2018 Mar 23.
Article in English | MEDLINE | ID: mdl-29570673

ABSTRACT

Activation of the hepatic cannabinoid type 1 receptor (CB1R) induces insulin resistance and gluconeogenesis via endoplasmic reticulum (ER) stress, thereby contributing to hyperglycemia. Gomisin N (GN) is a phytochemical derived from Schisandra chinensis. In the current study, we investigated the inhibitory effects of GN on hepatic CB1R-mediated insulin resistance and gluconeogenesis in 2-arachidonoylglycerol (AG; an agonist of CB1R)-treated HepG2 cells and in high-fat diet (HFD)-induced obese mice. Treatment with 2-AG induced the expression of ER stress markers, serine/threonine phosphatase PHLPP1, Lipin1, and ceramide synthesis genes, but reduced the expression of ceramide degradation genes in HepG2 cells. However, GN reversed 2-AG-mediated effects and improved the 2-AG-mediated impairment of insulin signaling. Furthermore, GN inhibited 2-AG-induced intracellular triglyceride accumulation and glucose production in HepG2 cells by downregulation of lipogenesis and gluconeogenesis genes, respectively. In vivo, GN administration to HFD obese mice reduced the HFD-induced increase in fasting blood glucose and insulin levels, which was accompanied with downregulation of HFD-induced expression of CB1R, ER stress markers, ceramide synthesis gene, and gluconeogenesis genes in the livers of HFD obese mice. These findings demonstrate that GN protects against hepatic CB1-mediated impairment of insulin signaling and gluconeogenesis, thereby contributing to the amelioration of hyperglycemia.


Subject(s)
Gluconeogenesis/drug effects , Lignans/pharmacology , Polycyclic Compounds/pharmacology , Receptors, Cannabinoid/metabolism , Arachidonic Acids/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Cyclooctanes/pharmacology , Endocannabinoids/pharmacology , Glycerides/pharmacology , Hep G2 Cells , Humans , Insulin Resistance/physiology , Lipogenesis/drug effects
13.
Biochem Biophys Res Commun ; 494(3-4): 587-593, 2017 12 16.
Article in English | MEDLINE | ID: mdl-29079190

ABSTRACT

Gomisin N (GN) is a lignan derived from Schisandra chinensis. AMP-activated kinase (AMPK) has gained attention as a therapeutic target for the treatment of metabolic syndrome. Previously, we reported that GN activated the AMPK pathway and ameliorated high-fat diet (HFD)-induced hepatic steatosis. In this study, we investigated the anti-diabetic effects of GN in C2C12 myotubes and HFD obese mice. GN enhanced the phosphorylation of AMPK/acetyl-CoA carboxylase (ACC) and Akt. In addition, GN promoted glucose uptake in C2C12 myotubes, which was accompanied by the translocation of glucose transporter 4 (GLUT4) to the plasma membrane. Treatment with compound C, an AMPK inhibitor, suppressed GN-mediated stimulation of glucose uptake. Furthermore, GN increased the expression of mitochondria biogenesis and fatty acid oxidation genes in C2C12 myotubes. In the in vivo study, administration of GN to HFD mice decreased the levels of fasting blood glucose and insulin, and improved glucose tolerance in HFD obese mice. GN administration rescued the decreased phosphorylation of AMPK and Akt and stimulated the expression of mitochondria biogenesis genes in the skeletal muscle of HFD mice. These findings suggested that GN exerted anti-hyperglycemic effects through AMPK activation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Blood Glucose/metabolism , Diabetes Mellitus/drug therapy , Diabetes Mellitus/enzymology , Lignans/administration & dosage , Mitochondria, Muscle/drug effects , Muscle, Skeletal/enzymology , Polycyclic Compounds/administration & dosage , Animals , Blood Glucose/drug effects , Cyclooctanes/administration & dosage , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Female , Hypoglycemic Agents/administration & dosage , Insulin/blood , Insulin Resistance , Mice , Mice, Inbred C57BL , Mitochondria, Muscle/metabolism , Muscle, Skeletal/drug effects , Organelle Biogenesis
14.
Pharmacogn Mag ; 13(49): 141-147, 2017.
Article in English | MEDLINE | ID: mdl-28216898

ABSTRACT

BACKGROUND: Daikenchuto (DKT; TJ-100, TU-100), a traditional herbal medicineis used in modern medicine to treat gastrointestinal (GI) functional disorders. Interstitial cells of Cajal (ICCs) are the pacemaker cells of the GI tract and play important roles in the regulation of GI motility. OBJECTIVE: The objective of this study was to investigate the effects of DKT on the pacemaker potentials (PPs) of cultured ICCs from murine small intestine. MATERIALS AND METHODS: Enzymatic digestions were used to dissociate ICCs from mouse small intestine tissues. All experiments on ICCs were performed after 12 h of culture. The whole-cell patch-clamp configuration was used to record ICC PPs (current clamp mode). All experiments were performed at 30-32°C. RESULTS: In current-clamp modeDKT depolarized and concentration-dependently decreased the amplitudes of PPs. Y25130 (a 5-HT3 receptor antagonist) or SB269970 (a 5-HT7 receptor antagonist) did not block DKT-induced PP depolarization, but RS39604 (a 5-HT4 receptor antagonist) did. Methoctramine (a muscarinic M2 receptor antagonist) failed to block DKT-induced PP depolarization, but pretreating 4-diphenylacetoxy-N-methylpiperidine methiodide (a muscarinic M3 receptor antagonist) facilitated blockade of DKT-induced PP depolarization. Pretreatment with an external Ca2+-free solution or thapsigargin abolished PPsand under these conditions, DKT did not induce PP depolarization. Furthermore Ginseng radix and Zingiberis rhizomes depolarized PPs, whereas Zanthoxyli fructus fruit (the third component of DKT) hyperpolarized PPs. CONCLUSION: These results suggest that DKT depolarizes ICC PPs in an internal or external Ca2+-dependent manner by stimulating 5-HT4 and M3 receptors. Furthermore, the authors suspect that the component in DKT largely responsible for depolarization is probably also a component of Ginseng radix and Zingiberis rhizomes. SUMMARY: Daikenchuto (DKT) depolarized and concentration-dependently decreased the amplitudes of pacemaker potentials (PPs)Y25130 (a 5-HT3 receptor antagonist) or SB269970 (a 5-HT7 receptor antagonist) did not block DKT-induced PP depolarization, but RS39604 (a 5-HT4 receptor antagonist) didMethoctramine (a muscarinic M2 receptor antagonist) failed to block DKT-induced PP depolarization, but pretreating 4-DAMP (a muscarinic M3 receptor antagonist) facilitated blockade of DKT-induced PP depolarizationGinseng radix and Zingiberis rhizomes depolarized PPswhereas Zanthoxyli fructus fruit (the third component of DKT) hyperpolarized PPs. Abbreviation used: DKT: Daikenchuto, GI: Gastrointestinal, ICCs: Interstitial cells of Cajal, PPs: Pacemaker Potentials.

15.
Sci Rep ; 7: 40345, 2017 01 09.
Article in English | MEDLINE | ID: mdl-28067305

ABSTRACT

Gomisin N (GN) is a physiological lignan derived from Schisandra chinensis. In the present study, we investigated the inhibitory effects of GN on differentiation of 3T3-L1 preadipocytes and the anti-obesity effects of GN in high-fat diet (HFD)-induced obese mice. Incubation with GN significantly inhibited the differentiation of 3T3-L1 preadipocytes in a dose-dependent manner. This inhibitory effect primarily occurred at an early adipogenic stage through impairment of mitotic clonal expansion (MCE) caused by cell cycle arrest at the G1/S phase transition. GN inhibited the extracellular signal-regulated kinase and phosphoinositide 3-kinase/protein kinase B signaling in the MCE process and activated AMP-activated protein kinase. Furthermore, GN downregulated CCAT/enhancer-binding protein ß (C/EBPß) and histone H3K9 demethylase JMJD2B during early stages of adipogenesis, and therefore repressed the expression of C/EBPß-targeted cell cycle genes. In addition, GN also repressed the expression of histone H3K4 methyltransferase MLL4 and reduced PPARγ expression. Moreover, GN effectively lowered the final body weight, adipose tissue mass, and reduced the serum levels of glucose, total triglyceride, and cholesterol in the HFD-induced obese mice. GN also markedly reduced hepatic triglyceride level induced by HFD. Collectively, these findings suggest that GN has potential as a novel agent for the prevention and treatment of obesity.


Subject(s)
Adipogenesis/drug effects , Lignans/pharmacology , Lignans/therapeutic use , Obesity/drug therapy , Obesity/prevention & control , Polycyclic Compounds/pharmacology , Polycyclic Compounds/therapeutic use , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Clone Cells , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 6/metabolism , Cyclins/metabolism , Cyclooctanes/pharmacology , Cyclooctanes/therapeutic use , Diet, High-Fat , Down-Regulation/drug effects , Fatty Liver/complications , Fatty Liver/drug therapy , Fatty Liver/pathology , Histone-Lysine N-Methyltransferase , Male , Mice , Mice, Inbred C57BL , Mitosis/drug effects , Obesity/blood , Obesity/complications , PPAR gamma/metabolism , Signal Transduction/drug effects
16.
PLoS One ; 12(1): e0168185, 2017.
Article in English | MEDLINE | ID: mdl-28060835

ABSTRACT

Previous studies have shown that tri- or di-methylation of histone H3 at lysine 9 (H3K9me3/me2) on the promoter of the peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein α (C/EBPα) contribute to the repression of PPARγ and C/EBPα and inhibition of adipogenesis in 3T3-L1 preadipocytes. The balance of histone methylation is regulated by histone methyltransferases and demethylases. However, it is poorly understood which demethylases are responsible for removing H3K9me3/me2 on the promoter of PPARγ and C/EBPα. JMJD2B is a H3K9me3/me2 demethylase that was previously shown to activate adipogenesis by promoting mitotic clonal expansion. Nevertheless, it remains unclear whether JMJD2B plays a role in the regulation of adipogenesis by removing H3K9me3/me2 on the promoter of PPARγ and C/EBPα and subsequently activating PPARγ and C/EBPα expression. Here, we showed that JMJD2B decreased H3K9me3/me2 on the promoter of PPARγ and C/EBPα, which in turn stimulated the expression of PPARγ and C/EBPα. JMJD2B knockdown using siRNA in 3T3-L1 preadipocytes repressed the expression of PPARγ and C/EBPα, resulting in inhibition of adipogenesis. This was accompanied by increased enrichment of H3K9me3/me2 on the promoter of PPARγ and C/EBPα. In contrast, overexpression of JMJD2B increased the expression of PPARγ and C/EBPα, which was accompanied by decreased enrichment of H3K9me3/me2 on the promoter and activated adipogenesis. Together, these results indicate that JMJD2B regulates PPARγ and C/EBPα during adipogenesis.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Gene Expression Regulation , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , PPAR gamma/genetics , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/genetics , Animals , Jumonji Domain-Containing Histone Demethylases/genetics , Methylation , Mice , Promoter Regions, Genetic
17.
Biochem Biophys Res Commun ; 482(4): 1095-1101, 2017 Jan 22.
Article in English | MEDLINE | ID: mdl-27914812

ABSTRACT

Gomisin N (GN) is a phytochemical derived from Schisandra chinensis. It has been reported to exert a protective effect against hepatic steatosis by attenuating endoplasmic reticulum (ER) stress. However, the detailed mechanism by which GN inhibits hepatic steatosis remains to be elucidated. In this study, we examined whether GN activates AMP-activated protein kinase (AMPK) and exerts therapeutic effects on liver X receptor (LXR)- or palmitic acid (PA)-induced triglyceride (TG) accumulation in HepG2 cells. Furthermore, in vivo protective effects of GN against hepatic steatosis were assessed in high-fat diet (HFD)-induced obese mice. GN stimulated the phosphorylation of AMPK, acetyl-CoA carboxylase (ACC), and sterol regulatory element-binding protein 1c (SREBP1c) in HepG2 cells. It decreased the expression of lipogenesis genes, but increased the expression of fatty acid oxidation genes. Additionally, GN decreased the expression of lipogenesis genes induced by the LXR agonist T0901317 or PA in HepG2 cells, resulting in reduced intracellular TG content. However, preincubation with compound C, an AMPK inhibitor, prevented GN-mediated effects. Administration of GN to HFD-induced obese mice decreased HFD-induced liver weight, hepatic TG accumulation, and cytoplasmic lipid droplet. These findings demonstrate that GN activates the AMPK pathway and ameliorates HFD-induced hepatic steatosis.


Subject(s)
Fatty Liver/metabolism , Lignans/pharmacology , Liver X Receptors/metabolism , Liver/metabolism , Polycyclic Compounds/pharmacology , Protein Kinases/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Anti-Inflammatory Agents/pharmacology , Cyclooctanes/pharmacology , Cytoplasm/metabolism , Fatty Acids/chemistry , Hep G2 Cells , Humans , Hydrocarbons, Fluorinated/pharmacology , Lipogenesis , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Phosphorylation , Phytochemicals/pharmacology , Sulfonamides/pharmacology , Triglycerides/chemistry
18.
PLoS One ; 11(9): e0162238, 2016.
Article in English | MEDLINE | ID: mdl-27611969

ABSTRACT

In this study, we performed high throughput RNA sequencing at the preadipocyte (D0) and differentiated adipocyte (D7) stages of primary brown preadipocyte differentiation in order to characterize the transcriptional events regulating differentiation and function. Compared to the preadipocyte stage (D0), 6,668 genes were identified as differentially expressed genes (DEGs) with a fold change of ≥ 2.0 at the differentiated adipocyte stage (D7). Several adipogenic genes including peroxisome proliferator-activated receptor-γ (PPARγ) and CCAAT/enhancer-binding protein-α (C/EBPα), and Krüppel-like factor (KLF) family genes were differentially expressed at D0 and D7. Since KLF16 gene expression was downregulated at day 7 and its adipogenic function has not been characterized, we investigated its role in adipogenesis. Knockdown of KLF16 stimulated the differentiation of both brown and 3T3-L1 preadipocytes, and led to increased PPARγ expression. However, overexpression of KLF16 had opposite effects. Furthermore, KLF16 downregulated PPARγ expression in brown adipocytes and inhibited its promoter activity. These results indicate that KLF16 inhibits adipogenesis through downregulation of PPARγ expression.


Subject(s)
Adipogenesis/physiology , Kruppel-Like Transcription Factors/physiology , Adipogenesis/genetics , Adipose Tissue, Brown/cytology , Animals , Female , Gene Expression Regulation , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL , PPAR gamma/genetics , PPAR gamma/metabolism , Sequence Analysis, RNA , Transcriptome
19.
Mol Med Rep ; 14(4): 3985-91, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27601272

ABSTRACT

Berberine is traditionally used to treat gastrointestinal (GI) motility disorders. The interstitial cells of Cajal (ICCs) are the pacemaker cells of the gastrointestinal tract, which are responsible for the production of gut movements. The present study aimed to investigate the effects of berberine on pacemaker potentials (PPs) in cultured ICC clusters from the mouse small intestine, and sought to identify the receptors involved and the underlying mechanisms of action. All experiments were performed on cultured ICCs, and a whole­cell patch­clamp configuration was used to record PPs from ICC clusters (current clamp mode). Under current clamp mode, berberine was shown to decrease the amplitude and frequency of PPs. However, these effects were suppressed by treatment with glibenclamide, a specific ATP­sensitive K+ channel blocker. Nor­binaltorphimine dihydrochloride (a kappa opioid receptor antagonist) did not suppress berberine­induced PP inhibition, whereas ICI 174,864 (a delta opioid receptor antagonist) and CTOP (a mu opioid receptor antagonist) did suppress the inhibitory effects of berberine. Pretreatment with SQ­22536 (an adenylate cyclase inhibitor) or with KT­5720 (a protein kinase A inhibitor) did not suppress the effects of berberine; however, pretreatment with 1H­[1,2,4] oxadiazolo [4,3­a] quinoxalin­1­one (a guanylate cyclase inhibitor) or KT­5823 [a protein kinase G (PKG) inhibitor] did. In addition, berberine stimulated cyclic guanosine monophosphate (cGMP) production in ICCs. These observations indicate that berberine may inhibit the pacemaker activity of ICC clusters via ATP­sensitive K+ channels and the cGMP­PKG­dependent pathway by stimulating mu and delta opioid receptors. Therefore, berberine may provide a basis for the development of novel agents for the treatment of GI motility dysfunction.


Subject(s)
Antidiarrheals/pharmacology , Berberine/pharmacology , Cyclic GMP/metabolism , Interstitial Cells of Cajal/drug effects , KATP Channels/metabolism , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Animals , Anti-Infective Agents/pharmacology , Berberine/chemistry , Berberis/chemistry , Cells, Cultured , Cyclic GMP-Dependent Protein Kinases/metabolism , Guanylate Cyclase/metabolism , Interstitial Cells of Cajal/cytology , Interstitial Cells of Cajal/metabolism , Intestine, Small/cytology , Intestine, Small/drug effects , Intestine, Small/metabolism , Mice , Mice, Inbred BALB C
20.
BMC Complement Altern Med ; 16: 187, 2016 Jul 07.
Article in English | MEDLINE | ID: mdl-27386946

ABSTRACT

BACKGROUND: Hominis Placenta (HP) known as a restorative medicine in Traditional Chinese Medicine (TCM), has been widely applied in the clinics of Korea and China as an anti-aging agent to enhance the regeneration of tissue. This study was conducted to investigate whether topical treatment of HP promotes hair regrowth in the animal model. METHODS: The dorsal hairs of 8-week-old C57BL/6 mice were depilated to synchronize hair follicles to the anagen phase. HP was applied topically once a day for 15 days. Hair growth was evaluated visually and microscopically. The incorporation of bromodeoxyuridine (BrdU) and expression of proliferating cell nuclear antigen (PCNA), fibroblast growth factor-7 (FGF-7) in dorsal skin tissue was examined by immunohistochemical analysis. Reverse transcription polymerase chain reaction (RT-PCR) was used to measure the mRNA expression of FGF-7. RESULTS: HP exhibited potent hair growth-promoting activity in C57BL/6 mice. Gross examination indicated that HP markedly increased hair regrowth as well as hair density and diameter. Histologic analysis showed that HP treatment enhanced the anagen induction of hair follicles. Immunohistochemical analysis revealed that BrdU incorporation and the expressions of PCNA were increased by treatment of HP. HP treatment significantly increased the expression of FGF-7, which plays pivotal roles to maintain anagen phase both protein and mRNA levels. CONCLUSIONS: Taken together, our results indicate that HP has a potent hair growth-promoting activity; therefore, it may be a good candidate for the treatment of alopecia.


Subject(s)
Biological Products/pharmacology , Hair Follicle/drug effects , Hair/drug effects , Medicine, Chinese Traditional , Placenta/chemistry , Animals , Back/physiology , Biological Products/chemistry , Bromodeoxyuridine/analysis , Bromodeoxyuridine/metabolism , Cell Proliferation/drug effects , Female , Fibroblast Growth Factor 7/analysis , Fibroblast Growth Factor 7/genetics , Fibroblast Growth Factor 7/metabolism , Hair Follicle/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...