Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
PLoS One ; 13(7): e0200336, 2018.
Article in English | MEDLINE | ID: mdl-29979770

ABSTRACT

Lobeglitazone (Lobe) is a novel thiazolidinedione antidiabetic drug that reduces insulin resistance by activating peroxisome proliferator-activated receptor-gamma (PPARγ). However, the exact mechanisms of antidiabetic effects of Lobe have not been established in an animal model. The aim of this study was to evaluate the hypoglycemic effects of Lobe and investigate possible factors involved in Lobe-enhanced hepatic steatosis in high-fat diet (HFD)-fed mice. Mice were fed an HFD for 15 weeks. Lobe was administrated orally during the last 9 weeks. Lobe treatment significantly reduced insulin resistance and increased expression of hepatic glucose transporter 4 (GLUT4) and PPARs in HFD-fed mice. However, increased body weight and hepatic steatosis were not reduced by Lobe in these mice. Metabolomics fingerprinting showed that several lipogenesis-related hepatic and serum metabolites in HFD-fed mice had positive or negative correlations with Lobe administration. In particular, increased leptin levels during HFD were further increased by Lobe. HFD-induced signaling transducer and activator of transcription 3 (STAT3) phosphorylation in the hypothalamus was increased by Lobe. In addition, immunohistochemical analysis showed more proopiomelanocortin (POMC)-positive neurons in the hypothalamus of HFD-fed mice (with or without Lobe) compared with normal diet-fed mice. Despite improving leptin signaling in the hypothalamus and enhancing insulin sensitivity in HFD-fed mice, Lobe increased body weight and steatosis. Further research is necessary regarding other factors affecting Lobe-enhanced hepatic steatosis and hyperphagia.


Subject(s)
Fatty Liver/drug therapy , Hypoglycemic Agents/pharmacology , Insulin Resistance/physiology , Liver/drug effects , PPAR gamma/agonists , Pyrimidines/pharmacology , Thiazolidinediones/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Diet, High-Fat , Fatty Liver/metabolism , Glucose Transporter Type 4/metabolism , Hypoglycemic Agents/therapeutic use , Lipogenesis/drug effects , Liver/metabolism , Mice , PPAR gamma/metabolism , Phosphorylation/drug effects , Pyrimidines/therapeutic use , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Thiazolidinediones/therapeutic use
2.
Korean J Physiol Pharmacol ; 22(3): 301-309, 2018 May.
Article in English | MEDLINE | ID: mdl-29719452

ABSTRACT

Statins mediate vascular protection and reduce the prevalence of cardiovascular diseases. Recent work indicates that statins have anticonvulsive effects in the brain; however, little is known about the precise mechanism for its protective effect in kainic acid (KA)-induced seizures. Here, we investigated the protective effects of atorvastatin pretreatment on KA-induced neuroinflammation and hippocampal cell death. Mice were treated via intragastric administration of atorvastatin for 7 days, injected with KA, and then sacrificed after 24 h. We observed that atorvastatin pretreatment reduced KA-induced seizure activity, hippocampal cell death, and neuroinflammation. Atorvastatin pretreatment also inhibited KA-induced lipocalin-2 expression in the hippocampus and attenuated KA-induced hippocampal cyclooxygenase-2 expression and glial activation. Moreover, AKT phosphorylation in KA-treated hippocampus was inhibited by atorvastatin pretreatment. These findings suggest that atorvastatin pretreatment may protect hippocampal neurons during seizures by controlling lipocalin-2-associated neuroinflammation.

3.
Biochem Biophys Res Commun ; 499(4): 1025-1031, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29634925

ABSTRACT

Chronic low-grade inflammation-induced insulin resistance is associated with neuroinflammation. Myeloid sirtuin1 (SIRT1) deficiency aggravates high-fat diet (HFD)-induced insulin resistance. However, the function of myeloid-specific SIRT1 in the hippocampus of obese mice is largely unknown. To address this question, we fed myeloid SIRT1 knockout (KO) mice a HFD for 40 weeks. We found that HFD-fed SIRT1 KO mice had increased insulin resistance and macrophage infiltration in adipose tissue than wild type (WT) mice. Levels of HFD-induced lipocalin-2 (LCN2) were lower in SIRT1 KO mice than in WT. HFD-induced hippocampal LCN2 expression was lower in HFD-fed SIRT1 KO mice than in WT. Hippocampal acetylation of nuclear factor-κB (NF-κB) and amyloid precursor protein levels were higher in HFD-fed SIRT1 KO mice than in HFD-fed WT mice. Taken together, our results suggest that targeted induction of the anti-inflammatory effects of SIRT1 and LCN2 may help prevent obesity-associated insulin resistance and neuroinflammation.


Subject(s)
Feeding Behavior , Hippocampus/pathology , Inflammation/pathology , Myeloid Cells/metabolism , Sirtuin 1/deficiency , Adipocytes/metabolism , Animals , Body Weight , Diet, High-Fat , Insulin Resistance , Lipocalin-2/blood , Macrophages/metabolism , Mice, Knockout , Sirtuin 1/metabolism
4.
Korean J Physiol Pharmacol ; 22(1): 63-70, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29302213

ABSTRACT

Cilostazol is a selective inhibitor of type 3 phosphodiesterase (PDE3) and has been widely used as an antiplatelet agent. Cilostazol mediates this activity through effects on the cyclic adenosine monophosphate (cAMP) signaling cascade. Recently, it has attracted attention as a neuroprotective agent. However, little is known about cilostazol's effect on excitotoxicity induced neuronal cell death. Therefore, this study evaluated the neuroprotective effect of cilostazol treatment against hippocampal neuronal damage in a mouse model of kainic acid (KA)-induced neuronal loss. Cilostazol pretreatment reduced KA-induced seizure scores and hippocampal neuron death. In addition, cilostazol pretreatment increased cAMP response element-binding protein (CREB) phosphorylation and decreased neuroinflammation. These observations suggest that cilostazol may have beneficial therapeutic effects on seizure activity and other neurological diseases associated with excitotoxicity.

5.
Sci Rep ; 7(1): 7837, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28798347

ABSTRACT

Recent studies have shown that overexpression of tonicity-responsive enhancer binding protein (TonEBP) is associated with many inflammatory diseases, including diabetes mellitus, which causes neuroinflammation in the hippocampus as well as hepatic steatosis. However, the exact mechanism in diabetic neuroinflammation is unknown. We report that haploinsufficiency of TonEBP inhibits hepatic and hippocampal high-mobility group box-1 (HMGB1) expression in diabetic mice. Here, mice were fed a high-fat diet (HFD) for 16 weeks and received an intraperitoneal injection of 100 mg/kg streptozotocin (STZ) and followed by continued HFD feeding for an additional 4 weeks to induce hyperglycemia and hepatic steatosis. Compared with wild-type diabetic mice, diabetic TonEBP+/- mice showed decreased body weight, fat mass, hepatic steatosis, and macrophage infiltration. We also found that adipogenesis and HMGB1 expression in the liver and hippocampus were lower in diabetic TonEBP+/- mice compared with the wild type. Furthermore, iba-1 immunoreactivity in the hippocampus was decreased in diabetic TonEBP+/- mice compared with that in the wild type. Our findings suggest that TonEBP haploinsufficiency suppresses diabetes-associated hepatic steatosis and neuroinflammation.


Subject(s)
Diabetes Mellitus, Experimental/complications , Encephalitis/pathology , Fatty Liver/complications , Haploinsufficiency , Hippocampus/pathology , Transcription Factors/genetics , Animals , Body Fat Distribution , Body Weight , Diet, High-Fat , Encephalitis/chemically induced , Gene Expression , HMGB1 Protein/biosynthesis , Mice
7.
Sci Rep ; 6: 30111, 2016 07 21.
Article in English | MEDLINE | ID: mdl-27439777

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is one of the most frequent causes of liver disease and its prevalence is a serious and growing clinical problem. Caloric restriction (CR) is commonly recommended for improvement of obesity-related diseases such as NAFLD. However, the effects of CR on hepatic metabolism remain unknown. We investigated the effects of CR on metabolic dysfunction in the liver of obese diabetic db/db mice. We found that CR of db/db mice reverted insulin resistance, hepatic steatosis, body weight and adiposity to those of db/m mice. (1)H-NMR- and UPLC-QTOF-MS-based metabolite profiling data showed significant metabolic alterations related to lipogenesis, ketogenesis, and inflammation in db/db mice. Moreover, western blot analysis showed that lipogenesis pathway enzymes in the liver of db/db mice were reduced by CR. In addition, CR reversed ketogenesis pathway enzymes and the enhanced autophagy, mitochondrial biogenesis, collagen deposition and endoplasmic reticulum stress in db/db mice. In particular, hepatic inflammation-related proteins including lipocalin-2 in db/db mice were attenuated by CR. Hepatic metabolomic studies yielded multiple pathological mechanisms of NAFLD. Also, these findings showed that CR has a therapeutic effect by attenuating the deleterious effects of obesity and diabetes-induced multiple complications.


Subject(s)
Body Weight , Caloric Restriction , Liver/metabolism , Non-alcoholic Fatty Liver Disease/diet therapy , Animals , Chromatography, Liquid , Collagen/metabolism , Endoplasmic Reticulum Stress , Ketones/metabolism , Lipid Metabolism , Lipogenesis , Mass Spectrometry , Metabolomics , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Proton Magnetic Resonance Spectroscopy , Triglycerides/biosynthesis
8.
Neurobiol Aging ; 44: 127-137, 2016 08.
Article in English | MEDLINE | ID: mdl-27318140

ABSTRACT

Diabetes may adversely affect cognitive function and, conversely, caloric restriction (CR) increases longevity and improves memory. To shed light on the unknown underlying mechanisms involved in these observations, we examined the effects of CR on serum metabolic parameters and hippocampal protein expression in the ob/ob mice model of obesity-induced diabetes. We found that CR reduced hepatic steatosis and insulin resistance in ob/ob mice. In addition, CR increased the levels of hippocampal O-linked-N-acetylglucosamine (O-GlcNAc) and GlcNAc transferase and decreased the expression of calcium/calmodulin-dependent protein kinase II, lipocalin-2, and phosphorylated tau. Furthermore, CR lessened the learning deficits that are typically seen in ob/ob mice. These findings indicate that CR may reverse obesity-related brain glucose impairment and intracellular Ca(2+) dysfunction and relieve learning impairment associated with diabetes.


Subject(s)
Acetylglucosamine/metabolism , Calcium Signaling/physiology , Caloric Restriction , Hippocampus/metabolism , Hippocampus/physiopathology , Learning Disabilities/etiology , Learning , Animals , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Diabetes Complications/complications , Fatty Liver/prevention & control , Insulin Resistance , Male , Mice, Inbred C57BL , Mice, Obese , N-Acetylglucosaminyltransferases/metabolism , Phosphorylation , tau Proteins/metabolism
9.
J Med Food ; 19(3): 290-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26741655

ABSTRACT

Artemisia annua L. (AA) is a well-known source of the antimalarial drug artemisinin. AA also has an antibacterial and antioxidant activity. However, the effect of AA extract on hepatic steatosis induced by obesity is unclear. We investigated whether AA extract prevents obesity-induced insulin resistance and hepatic steatosis in high-fat diet (HFD)-fed mice. Mice were randomly divided into groups that received a normal chow diet or HFD with or without AA for 12 weeks. We found that AA extract reduced insulin resistance and hepatic steatosis in HFD-fed mice. Western blot analysis showed that HFD-induced expression of nuclear sterol regulatory element-binding protein 1 and carbohydrate-responsive element-binding protein in the livers was decreased by AA extract. In particular, dietary administration of AA extract decreased hepatic high-mobility group box 1 and cyclooxygenase-2 expression in HFD-fed mice. AA extract also attenuated HFD-induced collagen deposition and fibrosis-related transforming growth factor-ß1 and connective tissue growth factor. These data indicate that dietary AA extract has beneficial effects on hepatic steatosis and inflammation in HFD-fed mice.


Subject(s)
Artemisia annua/chemistry , Fatty Liver/drug therapy , Plant Extracts/administration & dosage , Animals , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Diet, High-Fat/adverse effects , Fatty Liver/genetics , Fatty Liver/immunology , Humans , Male , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/genetics , Obesity/immunology , Plant Leaves/chemistry , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/immunology
10.
J Cereb Blood Flow Metab ; 36(6): 1098-110, 2016 06.
Article in English | MEDLINE | ID: mdl-26661177

ABSTRACT

Diabetes-induced cognitive decline has been recognized in human patients of type 2 diabetes mellitus and mouse model of obesity, but the underlying mechanisms or therapeutic targets are not clearly identified. We investigated the effect of caloric restriction on diabetes-induced memory deficits and searched a molecular mechanism of caloric restriction-mediated neuroprotection. C57BL/6 mice were fed a high-fat diet for 40 weeks and RNA-seq analysis was performed in the hippocampus of high-fat diet-fed mice. To investigate caloric restriction effect on differential expression of genes, mice were fed high-fat diet for 20 weeks and continued on high-fat diet or subjected to caloric restriction (2 g/day) for 12 weeks. High-fat diet-fed mice exhibited insulin resistance, glial activation, blood-brain barrier leakage, and memory deficits, in that we identified neurogranin, a down-regulated gene in high-fat diet-fed mice using RNA-seq analysis; neurogranin regulates Ca(2+)/calmodulin-dependent synaptic function. Caloric restriction increased insulin sensitivity, reduced high-fat diet-induced blood-brain barrier leakage and glial activation, and improved memory deficit. Furthermore, caloric restriction reversed high-fat diet-induced expression of neurogranin and the activation of Ca(2+)/calmodulin-dependent protein kinase II and calpain as well as the downstream effectors. Our results suggest that neurogranin is an important factor of high-fat diet-induced memory deficits on which caloric restriction has a therapeutic effect by regulating neurogranin-associated calcium signaling.


Subject(s)
Calcium Signaling , Caloric Restriction , Cognition Disorders/diet therapy , Diabetes Mellitus, Type 2/complications , Neurogranin/genetics , Animals , Cognition Disorders/etiology , Diet, High-Fat , Gene Expression Profiling , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Sequence Analysis, RNA
11.
BMC Neurosci ; 16: 72, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26518260

ABSTRACT

BACKGROUND: Obesity has deleterious effects on the brain, and metabolic dysfunction may exacerbate the outcomes of seizures and brain injuries. However, it is unclear whether obesity affects excitotoxicity-induced neuronal cell death. The purpose of this study was to investigate the effects of a high-fat diet (HFD) on neuroinflammation and oxidative stress in the hippocampus of kainic acid (KA)-treated mice. RESULTS: Mice were fed with a HFD or normal diet for 8 weeks and then received a systemic injection of KA. HFD-fed mice showed hypercholesterolemia, insulin resistance, and hepatic steatosis. HFD-fed mice showed greater susceptibility to KA-induced seizures, an increased number of terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cells, neuroinflammation, and oxidative stress. Furthermore, we found that KA treatment increased HFD-induced calpain1, nuclear factor E2-related factor 2, and heme oxygenase-1 expression in the hippocampus. CONCLUSIONS: These findings imply that complex mechanisms affected by obesity-induced systemic inflammation, neuroinflammation, ER stress, calcium overload, and oxidative stress may contribute to neuronal death after brain injury.


Subject(s)
Cell Death/physiology , Diet, High-Fat , Excitatory Amino Acid Agonists/pharmacology , Hippocampus/metabolism , Kainic Acid/pharmacology , Neurons/metabolism , Obesity/complications , Animals , Cell Death/drug effects , Fatty Liver/etiology , Hippocampus/drug effects , Hypercholesterolemia/etiology , In Situ Nick-End Labeling , Inflammation/etiology , Insulin Resistance/physiology , Male , Mice , Mice, Inbred ICR , Neurons/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Seizures/chemically induced
12.
Korean J Physiol Pharmacol ; 19(5): 451-60, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26330758

ABSTRACT

Sirtuin 1 (SIRT1) is a mammalian NAD(+)-dependent protein deacetylase that regulates cellular metabolism and inflammatory response. The organ-specific deletion of SIRT1 induces local inflammation and insulin resistance in dietary and genetic obesity. Macrophage-mediated inflammation contributes to insulin resistance and metabolic syndrome, however, the macrophage-specific SIRT1 function in the context of obesity is largely unknown. C57/BL6 wild type (WT) or myeloid-specific SIRT1 knockout (KO) mice were fed a high-fat diet (HFD) or normal diet (ND) for 12 weeks. Metabolic parameters and markers of hepatic steatosis and inflammation in liver were compared in WT and KO mice. SIRT1 deletion enhanced HFD-induced changes on body and liver weight gain, and increased glucose and insulin resistance. In liver, SIRT1 deletion increased the acetylation, and enhanced HFD-induced nuclear translocation of nuclear factor kappa B (NF-κB), hepatic inflammation and macrophage infiltration. HFD-fed KO mice showed severe hepatic steatosis by activating lipogenic pathway through sterol regulatory element-binding protein 1 (SREBP-1), and hepatic fibrogenesis, as indicated by induction of connective tissue growth factor (CTGF), alpha-smooth muscle actin (α-SMA), and collagen secretion. Myeloid-specific deletion of SIRT1 stimulates obesity-induced inflammation and increases the risk of hepatic fibrosis. Targeted induction of macrophage SIRT1 may be a good therapy for alleviating inflammation-associated metabolic syndrome.

13.
Free Radic Biol Med ; 89: 54-61, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26191652

ABSTRACT

Doxorubicin (Dox) is an effective anti-cancer drug, but its use is limited because of its adverse effect of inducing irreversible dilated cardiomyopathy. Cilostazol (Cilo), a potent phosphodiesterase III inhibitor, has been reported to have an anti-inflammatory effect. Here, we investigated whether Cilo has a protective effect against Dox-induced cardiomyopathy (DIC). Mice were randomly divided into four groups: saline control, Dox (15 mg/kg), Dox (15 mg/kg) plus Cilo (50mg/kg), and Cilo (50mg/kg). The results showed that the coadministration of Dox and Cilo significantly enhanced left-ventricular systolic function compared with Dox alone. In addition, Cilo treatment significantly reduced Dox-induced perivascular fibrosis, collagen concentration, and connective growth factor expression in the heart. Also, Cilo administration markedly reduced Dox-induced levels of serum B-type natriuretic peptide, dysferlin, high-mobility group protein B1, Toll-like receptor 4, nuclear factor-κB p65, and cyclooxygenase-2. Furthermore, Cilo treatment significantly reduced Dox-induced oxidative stress by lowering the translocation of Nrf2 into the nucleus and the expression of NQO1, heme oxygenase 1, and superoxide dismutase-1. Our results suggest that Cilo may be a potential antifibrotic, antioxidative, and anti-inflammatory drug for DIC.


Subject(s)
Cardiomyopathies/prevention & control , Doxorubicin/toxicity , Fibrosis/prevention & control , Inflammation/prevention & control , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Tetrazoles/pharmacology , Animals , Antibiotics, Antineoplastic/toxicity , Blotting, Western , Cardiomyopathies/chemically induced , Cardiomyopathies/pathology , Cilostazol , Echocardiography , Fibrosis/chemically induced , Fibrosis/pathology , Inflammation/chemically induced , Inflammation/pathology , Male , Mice , Mice, Inbred ICR
14.
PLoS One ; 10(6): e0131671, 2015.
Article in English | MEDLINE | ID: mdl-26114656

ABSTRACT

Radiation-induced lung injury (RILI) is a common and unavoidable complication of thoracic radiotherapy. The current study was conducted to evaluate the ability of clarithromycin (CLA) to prevent radiation-induced pneumonitis, oxidative stress, and lung fibrosis in an animal model. C57BL/6J mice were assigned to control, irradiation only, irradiation plus CLA, and CLA only groups. Test mice received single thoracic exposures to radiation and/or oral CLA (100 mg/kg/day). Histopathologic findings and markers of inflammation, fibrosis, and oxidative stress were compared by group. On a microscopic level, CLA inhibited macrophage influx, alveolar fibrosis, parenchymal collapse, consolidation, and epithelial cell changes. The concentration of collagen in lung tissue was lower in irradiation plus CLA mice. Radiation-induced expression of tumor necrosis factor (TNF)-α, TNF receptor 1, acetylated nuclear factor kappa B, cyclooxygenase 2, vascular cell adhesion molecule 1, and matrix metallopeptidase 9 were also attenuated by CLA. Expression levels of nuclear factor erythroid 2-related factor 2 and heme oxygenase 1, transforming growth factor-ß1, connective tissue growth factor, and type I collagen in radiation-treated lungs were also attenuated by CLA. These findings indicate that CLA ameliorates the deleterious effects of thoracic irradiation in mice by reducing pulmonary inflammation, oxidative damage, and fibrosis.


Subject(s)
Clarithromycin/administration & dosage , Radiation Pneumonitis/prevention & control , Radiation-Protective Agents/administration & dosage , Animals , Apoptosis/drug effects , Body Weight/drug effects , Body Weight/radiation effects , Disease Models, Animal , Female , Fibrosis , Lung/drug effects , Lung/pathology , Lung/radiation effects , Mice , Mice, Inbred C57BL , Radiation Pneumonitis/mortality , Radiation Pneumonitis/pathology
15.
Korean J Physiol Pharmacol ; 18(4): 333-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25177166

ABSTRACT

Exendin-4 (Ex-4), a glucagon-like peptide-1 receptor (GLP-1R) agonist, has been known to reverse hepatic steatosis in ob/ob mice. Although many studies have evaluated molecular targets of Ex-4, its mechanism of action on hepatic steatosis and fibrosis has not fully been determined. In the liver, glucose transporter 4 (GLUT4) is mainly expressed in hepatocytes, endothelial cells and hepatic stellate cells (HSCs). In the present study, the effects of Ex-4 on GLUT4 expression were determined in the liver of ob/ob mice. Ob/ob mice were treated with Ex-4 for 10 weeks. Serum metabolic parameters, hepatic triglyceride levels, and liver tissues were evaluated for hepatic steatosis. The weights of the whole body and liver in ob/ob mice were reduced by long-term Ex-4 treatment. Serum metabolic parameters, hepatic steatosis, and hepatic fibrosis in ob/ob mice were reduced by Ex-4. Particularly, Ex-4 improved hepatic steatosis by enhancing GLUT4 via GLP-1R activation in ob/ob mice. Ex-4 treatment also inhibited hepatic fibrosis by decreasing expression of connective tissue growth factor in HSCs of ob/ob mice. Our data suggest that GLP-1 agonists exert a protective effect on hepatic steatosis and fibrosis in obesity and type 2 diabetes.

16.
Biosci Biotechnol Biochem ; 78(3): 482-9, 2014.
Article in English | MEDLINE | ID: mdl-25036839

ABSTRACT

A Vigna nakashimae (VN) extract has been shown to have antidiabetic and anti-obesity effects. However, the mechanism underlying the effect of a VN extract on hepatic inflammation and endoplasmic reticulum (ER) stress remains unclear. In the present study, we investigated how a VN extract protects against the development of non-alcoholic fatty liver disease (NAFLD). A VN extract for 12 weeks reduced the body weight, serum metabolic parameters, cytokines, and hepatic steatosis in high-fat diet (HFD)-fed mice. A VN extract decreased HFD-induced hepatic acetyl CoA carboxylase and glucose transporter 4 expressions. In addition to the levels of high-mobility group box 1 and receptor for advanced glycation, the hepatic expression of ATF4 and caspase-3 was also reduced by a VN extract. Thus, these data indicate that a chronic VN extract prevented NAFLD through multiple mechanisms, including inflammation, ER stress, and apoptosis in the liver.


Subject(s)
Fabaceae/chemistry , Inflammation/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Plant Extracts/administration & dosage , Acetyl-CoA Carboxylase/biosynthesis , Animals , Apoptosis/drug effects , Diet, High-Fat , Gene Expression Regulation/drug effects , Glucose Transporter Type 4/biosynthesis , Humans , Inflammation/metabolism , Inflammation/pathology , Lipid Metabolism/drug effects , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Plant Extracts/chemistry
17.
Metab Brain Dis ; 29(3): 635-43, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24756314

ABSTRACT

Obesity-induced fatty liver disease is associated with increased hypothalamic inflammation. Previous reports have demonstrated that the deletion of SIRT1 in hepatocytes increases hepatic steatosis and inflammation. Using myeloid cell-specific SIRT1 knockout (KO) mice, we investigated whether ablation of SIRT1 in macrophages plays a role in regulating hepatic steatosis and hypothalamic inflammation. When challenged with a high-fat diet (HFD) for 24 weeks, hyperleptinemia, hyperinsulinemia, hepatic steatosis and macrophage infiltrations in HFD-fed KO mice were increased compared with HFD-fed WT mice. Hypothalamic expression levels of iba1 were increased in HFD-fed KO mice compared with HFD-fed WT mice. In particular, the expression levels of choline acetyltransferase were decreased in the hypothalamus of HFD-fed KO mice compared with HFD-fed WT mice. Thus, our findings suggest that SIRT1 plays a key role for hepatic steatosis and hypothalamic inflammation and that anti-inflammatory effect of SIRT1 may be important for the prevention of obesity-induced metabolic syndromes.


Subject(s)
Fatty Liver/metabolism , Hypothalamus/metabolism , Inflammation/metabolism , Myeloid Cells/metabolism , Sirtuin 1/metabolism , Animals , Diet, High-Fat , Fatty Liver/genetics , Fatty Liver/pathology , Glucose Tolerance Test , Hypothalamus/pathology , Inflammation/genetics , Inflammation/pathology , Insulin/blood , Insulin Resistance/genetics , Leptin/blood , Male , Mice , Mice, Knockout , Myeloid Cells/pathology , Sirtuin 1/genetics
18.
J Neural Transm (Vienna) ; 121(10): 1233-43, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24728926

ABSTRACT

Kainic acid (KA)-induced neuronal death is closely linked to endoplasmic reticulum (ER) and mitochondrial dysfunction. Parkin is an ubiquitin E3 ligase that mediates the ubiquitination of the Bcl-2 family of proteins and its mutations are associated with neuronal apoptosis in neurodegenerative diseases. We investigated the effect of salubrinal, an ER stress inhibitor, on the regulation of ER stress and mitochondrial apoptosis induced by KA, in particular, by controlling parkin expression. We showed that salubrinal significantly reduced seizure activity and increased survival rates of mice with KA-induced seizures. We found that salubrinal protected neurons against apoptotic death by reducing expression of mitochondrial apoptotic factors and elF2α-ATF4-CHOP signaling proteins. Interestingly, we showed that salubrinal decreased the KA-induced parkin expression and inhibited parkin translocation to mitochondria, which suggests that parkin may regulate a cross-talk between ER and mitochondria. Collectively, inhibition of ER stress attenuates mitochondrial apoptotic and ER stress pathways and controls parkin-mediated neuronal death following KA-induced seizures.


Subject(s)
Apoptosis/drug effects , Cinnamates/pharmacology , Endoplasmic Reticulum Stress/drug effects , Hippocampus/drug effects , Kainic Acid/toxicity , Neuroprotective Agents/pharmacology , Thiourea/analogs & derivatives , Animals , Anticonvulsants/pharmacology , Apoptosis/physiology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum/physiology , Endoplasmic Reticulum Stress/physiology , Hippocampus/pathology , Hippocampus/physiopathology , Male , Mice, Inbred ICR , Mitochondria/drug effects , Mitochondria/pathology , Mitochondria/physiology , Neurons/drug effects , Neurons/pathology , Neurons/physiology , Nitric Oxide Synthase Type II/metabolism , Random Allocation , Seizures/chemically induced , Seizures/drug therapy , Seizures/pathology , Seizures/physiopathology , Survival Analysis , Thiourea/pharmacology , Ubiquitin-Protein Ligases/metabolism
19.
Korean J Physiol Pharmacol ; 17(4): 267-74, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23946685

ABSTRACT

A beneficial radioprotective agent has been used to treat the radiation-induced lung injury. This study was performed to investigate whether curcumin, which is known to have anti-inflammatory and antioxidant properties, could ameliorate radiation-induced pulmonary inflammation and fibrosis in irradiated lungs. Rats were given daily doses of intragastric curcumin (200 mg/kg) prior to a single irradiation and for 8 weeks after radiation. Histopathologic findings demonstrated that macrophage accumulation, interstitial edema, alveolar septal thickness, perivascular fibrosis, and collapse in radiation-treated lungs were inhibited by curcumin administration. Radiation-induced transforming growth factor-ß1 (TGF-ß1), connective tissue growth factor (CTGF) expression, and collagen accumulation were also inhibited by curcumin. Moreover, western blot analysis revealed that curcumin lowered radiation-induced increases of tumor necrosis factor-α (TNF-α), TNF receptor 1 (TNFR1), and cyclooxygenase-2 (COX-2). Curcumin also inhibited the nuclear translocation of nuclear factor-κ B (NF-κB) p65 in radiation-treated lungs. These results indicate that long-term curcumin administration may reduce lung inflammation and fibrosis caused by radiation treatment.

20.
Cardiovasc Diabetol ; 11: 111, 2012 Sep 19.
Article in English | MEDLINE | ID: mdl-22992429

ABSTRACT

BACKGROUND: Hyperglycemia leads to cardiac oxidative stress and an imbalance in glucose homeostasis. Diabetic cardiomyopathy is characterised by cardiac hypertrophy and fibrosis. However, the underlying mechanisms of diabetic cardiomyopathy are not fully understood. This study aimed to investigate the effects of alpha-lipoic acid (ALA) on cardiac energy metabolism, antioxidant effect, and fibrosis in the hearts of Otsuka Long-Evans Tokushima fatty (OLETF) rats. METHODS: Animals were separated into non-diabetic Long-Evans Tokushima Otsuka (LETO) rats and diabetes-prone OLETF rats with or without ALA (200 mg/kg/day) administration for 16 weeks. Diabetic cardiomyopathy was assessed by staining with Sirius Red. The effect of ALA on AMPK signalling, antioxidant enzymes, and fibrosis-related genes in the heart of OLETF rats were performed by Western blot analysis or immunohistochemistry. RESULTS: Western blot analysis showed that cardiac adenosine monophosphate-activated kinase (AMPK) signalling was lower in OLETF rats than in LETO rats, and that ALA treatment increased the signalling in OLETF rats. Furthermore, the low antioxidant activity in OLETF rats was increased by ALA treatment. In addition to increased Sirius red staining of collagen deposits, transforming growth factor-ß1 (TGF-ß1) and connective tissue growth factor (CTGF) were expressed at higher levels in OLETF rat hearts than in LETO rat hearts, and the levels of these factors were decreased by ALA. CONCLUSIONS: ALA enhances AMPK signalling, antioxidant, and antifibrogenic effect. Theses findings suggest that ALA may have beneficial effects in the treatment of diabetic cardiomyopathy.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetic Cardiomyopathies/drug therapy , Thioctic Acid/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Diabetes Mellitus, Type 2/metabolism , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Energy Metabolism/drug effects , Fibrosis , Male , Rats , Rats, Inbred OLETF , Rats, Long-Evans , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...