Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
FASEB J ; 33(6): 7563-7577, 2019 06.
Article in English | MEDLINE | ID: mdl-30870003

ABSTRACT

Dietary inorganic nitrate prevents aspects of cardiac mitochondrial dysfunction induced by hypoxia, although the mechanism is not completely understood. In both heart and skeletal muscle, nitrate increases fatty acid oxidation capacity, and in the latter case, this involves up-regulation of peroxisome proliferator-activated receptor (PPAR)α expression. Here, we investigated whether dietary nitrate modifies mitochondrial function in the hypoxic heart in a PPARα-dependent manner. Wild-type (WT) mice and mice without PPARα (Ppara-/-) were given water containing 0.7 mM NaCl (control) or 0.7 mM NaNO3 for 35 d. After 7 d, mice were exposed to normoxia or hypoxia (10% O2) for the remainder of the study. Mitochondrial respiratory function and metabolism were assessed in saponin-permeabilized cardiac muscle fibers. Environmental hypoxia suppressed mass-specific mitochondrial respiration and additionally lowered the proportion of respiration supported by fatty acid oxidation by 18% (P < 0.001). This switch away from fatty acid oxidation was reversed by nitrate treatment in hypoxic WT but not Ppara-/- mice, indicating a PPARα-dependent effect. Hypoxia increased hexokinase activity by 33% in all mice, whereas lactate dehydrogenase activity increased by 71% in hypoxic WT but not Ppara-/- mice. Our findings indicate that PPARα plays a key role in mediating cardiac metabolic remodeling in response to both hypoxia and dietary nitrate supplementation.-Horscroft, J. A., O'Brien, K. A., Clark, A. D., Lindsay, R. T., Steel, A. S., Procter, N. E. K., Devaux, J., Frenneaux, M., Harridge, S. D. R., Murray, A. J. Inorganic nitrate, hypoxia, and the regulation of cardiac mitochondrial respiration-probing the role of PPARα.


Subject(s)
Cell Respiration , Hypoxia/metabolism , Mitochondria, Heart/metabolism , Nitrates/metabolism , PPAR alpha/physiology , Animals , Inorganic Chemicals/administration & dosage , Inorganic Chemicals/metabolism , Mice , Mice, Knockout , Myocardium/metabolism , Nitrates/administration & dosage , Oxidative Phosphorylation , PPAR alpha/genetics
2.
J Proteome Res ; 17(5): 1887-1897, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29664296

ABSTRACT

Nutmeg is a Traditional Chinese Medicine used to treat gastrointestinal diseases. Some reports have indicated that nutmeg has hepatoprotective activity. In this study, a thioacetamide (TAA)-induced acute liver injury model in mice was used to explore the mechanism of the protective effects of nutmeg extract (NME), including its major bioactive component myrislignan. The results indicated that NME could effectively protect TAA-induced liver damage as assessed by recovery of increased serumtransaminases, decrease in hepatic oxidative stress, and lower hepatic inflammation. Metabolomics analysis further revealed that treatment with NME led to the recovery of a series of lipids including lysophosphatidylcholines that were decreased and a lowering of acylcarnitines that were increased in mouse plasma and liver after TAA exposure. Gene expression analysis demonstrated that the hepatoprotective effect of NME was achieved by modulation of the peroxisome proliferator-activated receptor alpha (PPARα) as well as the decrease in oxidative stress. NME could not protect from TAA-induced liver injury in Ppara-null mice, suggesting that its protective effect was dependent on PPARα. Myrislignan, a representative neolignan in nutmeg, showed potent protective activity against TAA-induced liver toxicity. These data demonstrate that nutmeg alleviates TAA-induced liver injury through the modulation of PPARα and that the lignan compounds in nutmeg such as myrislignan partly contributed to this action.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Myristica , PPAR alpha/physiology , Animals , Carnitine/analogs & derivatives , Carnitine/analysis , Lipids/analysis , Metabolomics , Mice , Mice, Knockout , Oxidative Stress , Protective Agents/pharmacology , Thioacetamide/adverse effects
3.
Nutrients ; 8(12)2016 Dec 12.
Article in English | MEDLINE | ID: mdl-27973445

ABSTRACT

We previously reported that bitter melon seed oil (BMSO) was an effective anti-steatosis and antiobesity agent. Since the major fatty acid α-eleostearic acid (α-ESA) in BMSO is a peroxisome proliferator-activated receptor α (PPARα) activator, the objective was to investigate the role of PPARα in BMSO-modulated lipid disorders and α-ESA metabolism. C57BL/6J wild (WD) and PPARα knockout (KO) mice were fed a high-fat diet containing BMSO (15% soybean oil + 15% BMSO, HB) or not (30% soybean oil, HS) for 5 weeks. The HB diet significantly reduced hepatic triglyceride concentrations and increased acyl-CoA oxidase activity in WD, but not in KO mice. However, regardless of genotype, body fat percentage was lowered along with upregulated protein levels of uncoupling protein 1 (UCP1) and tyrosine hydroxylase, as well as signaling pathway of cAMP-dependent protein kinase and AMP-activated protein kinase in the white adipose tissue of HB-treated groups compared to HS cohorts. In WD-HB and KO-HB groups, white adipose tissue had autophagy, apoptosis, inflammation, and browning characteristics. Without PPARα, in vivo reduction of α-ESA into rumenic acid was slightly but significantly lowered, along with remarkable reduction of hepatic retinol saturase (RetSat) expression. We concluded that BMSO-mediated anti-steatosis depended on PPARα, whereas the anti-adiposity effect was PPARα-independent. In addition, PPARα-dependent enzymes may participate in α-ESA conversion, but only have a minor role.


Subject(s)
Dyslipidemias/drug therapy , Linoleic Acids, Conjugated/metabolism , Linolenic Acids/metabolism , Momordica charantia/chemistry , PPAR alpha/physiology , Phytotherapy , Plant Oils/chemistry , Acyl-CoA Oxidase/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adiposity/drug effects , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Diet, High-Fat/adverse effects , Dyslipidemias/metabolism , Fatty Liver/drug therapy , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidoreductases Acting on CH-CH Group Donors/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism , Plant Oils/administration & dosage , Signal Transduction/drug effects , Triglycerides/metabolism , Tyrosine 3-Monooxygenase/metabolism , Uncoupling Protein 1/metabolism
4.
Circ Res ; 117(1): 41-51, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25977309

ABSTRACT

RATIONALE: Post-ischemic contractile dysfunction is a contributor to morbidity and mortality after the surgical correction of congenital heart defects in neonatal patients. Pre-existing hypertrophy in the newborn heart can exacerbate these ischemic injuries, which may partly be due to a decreased energy supply to the heart resulting from low fatty acid ß-oxidation rates. OBJECTIVE: We determined whether stimulating fatty acid ß-oxidation with GW7647, a peroxisome proliferator-activated receptor-α (PPARα) activator, would improve cardiac energy production and post-ischemic functional recovery in neonatal rabbit hearts subjected to volume overload-induced cardiac hypertrophy. METHODS AND RESULTS: Volume-overload cardiac hypertrophy was produced in 7-day-old rabbits via an aorto-caval shunt, after which, the rabbits were treated with or without GW7647 (3 mg/kg per day) for 14 days. Biventricular working hearts were subjected to 35 minutes of aerobic perfusion, 25 minutes of global no-flow ischemia, and 30 minutes of aerobic reperfusion. GW7647 treatment did not prevent the development of cardiac hypertrophy, but did prevent the decline in left ventricular ejection fraction in vivo. GW7647 treatment increased cardiac fatty acid ß-oxidation rates before and after ischemia, which resulted in a significant increase in overall ATP production and an improved in vitro post-ischemic functional recovery. A decrease in post-ischemic proton production and endoplasmic reticulum stress, as well as an activation of sarcoplasmic reticulum calcium ATPase isoform 2 and citrate synthase, was evident in GW7647-treated hearts. CONCLUSIONS: Stimulating fatty acid ß-oxidation in neonatal hearts may present a novel cardioprotective intervention to limit post-ischemic contractile dysfunction.


Subject(s)
Butyrates/therapeutic use , Cardiomegaly/physiopathology , Myocardial Contraction/physiology , Myocardial Ischemia/drug therapy , Myocardium/metabolism , PPAR alpha/agonists , Phenylurea Compounds/therapeutic use , ATP Citrate (pro-S)-Lyase/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Animals, Newborn , Butyrates/pharmacology , Calcium-Transporting ATPases/metabolism , Cardiomegaly/prevention & control , Citric Acid Cycle/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Endoplasmic Reticulum Stress/drug effects , Enzyme Activation/drug effects , Fatty Acids/metabolism , Female , Glycolysis , Heart/drug effects , Inflammation , Male , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Myocardial Contraction/drug effects , PPAR alpha/physiology , Phenylurea Compounds/pharmacology , Rabbits , Sarcoplasmic Reticulum/enzymology , Stroke Volume/drug effects
5.
Alcohol ; 48(7): 707-15, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25262573

ABSTRACT

This study investigated the effects of umbelliferone (UF) on alcoholic fatty liver and its underlying mechanism. Rats were fed a Lieber-DeCarli liquid diet with 36% of calories as alcohol with or without UF (0.05 g/L) for 8 weeks. Pair-fed rats received an isocaloric carbohydrate liquid diet. UF significantly reduced the severity of alcohol-induced body weight loss, hepatic lipid accumulation and droplet formation, and dyslipidemia. UF decreased plasma AST, ALT, and γGTP activity. UF significantly reduced hepatic cytochrome P450 2E1 activities and increased alcohol dehydrogenase and aldehyde dehydrogenase 2 activities compared to the alcohol control group, which resulted in a lower plasma acetaldehyde level in the rats that received UF. Chronic alcohol exposure inhibited hepatic AMPK activation compared to the pair-fed rats, which was reversed by UF supplementation. UF also significantly suppressed the lipogenic gene expression (SREBP-1c, SREBP-2, FAS, CIDEA, and PPARγ) and elevated the fatty acid oxidation gene expression (PPARα, Acsl1, CPT, Acox, and Acaa1a) compared to the alcohol control group, which could lead to inhibition of FAS activity and stimulation of CPT and fatty acid ß-oxidation activities in the liver of chronic alcohol-fed rats. These results indicated that UF attenuated alcoholic steatosis through down-regulation of SREBP-1c-mediated lipogenesis and up-regulation of PPARα-mediated fatty acid oxidation. Therefore, UF may provide a promising natural therapeutic strategy against alcoholic fatty liver.


Subject(s)
Fatty Liver, Alcoholic/drug therapy , PPAR alpha/drug effects , Sterol Regulatory Element Binding Protein 1/drug effects , Umbelliferones/therapeutic use , Alcohol Dehydrogenase/metabolism , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase, Mitochondrial , Animals , Cytochrome P-450 CYP2E1/metabolism , Dietary Supplements , Hypolipidemic Agents/therapeutic use , Ifosfamide/analogs & derivatives , Ifosfamide/blood , Liver/drug effects , Liver/enzymology , Male , Mitochondrial Proteins/metabolism , PPAR alpha/physiology , Rats , Rats, Sprague-Dawley , Sterol Regulatory Element Binding Protein 1/physiology
6.
J Nutr Biochem ; 24(10): 1678-90, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23643524

ABSTRACT

Animal models are an important tool to understand the complex pathogenesis of inflammatory bowel diseases (IBDs). This study tested the anti-inflammatory potential of a green tea extract rich in polyphenols (GrTP) in the colon of the multidrug resistance targeted mutation (Mdr1a(-/-)) mouse model of IBD. Insights into mechanisms responsible for this reduction in inflammation were gained using transcriptome and proteome analyses. Mice were randomly assigned to an AIN-76A (control) or GrTP-enriched diet. At 21 or 24 weeks of age, a colonic histological injury score was determined for each mouse, colon mRNA transcript levels were assessed using microarrays, and colon protein expression was measured using two-dimensional gel electrophoresis and liquid chromatography-mass spectrometry protein identification. Mean colonic histological injury score of GrTP-fed Mdr1a(-/-) mice was significantly lower compared to those fed the control diet. Microarray and proteomics analyses showed reduced abundance of transcripts and proteins associated with immune and inflammatory response and fibrinogenesis pathways, and increased abundance of those associated with xenobiotic metabolism pathways in response to GrTP, suggesting that its anti-inflammatory activity is mediated by multiple molecular pathways. Peroxisome proliferator-activated receptor-α and signal transducer and activator of transcription 1 appear to be two key molecules which regulate these effects. These results support the view that dietary intake of polyphenols derived from green tea can ameliorate intestinal inflammation in the colon of a mouse model of IBD, and are in agreement with studies suggesting that consumption of green tea may reduce IBD symptoms and therefore play a part in an overall IBD treatment regimen.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/deficiency , Colitis/prevention & control , Colon/metabolism , Polyphenols/pharmacology , Animals , Colitis/pathology , Colon/drug effects , Colon/pathology , Inflammatory Bowel Diseases/prevention & control , Male , Mice , Models, Animal , PPAR alpha/physiology , Proteome , STAT1 Transcription Factor/physiology , Tea/chemistry , Transcriptome
7.
Am J Physiol Gastrointest Liver Physiol ; 304(1): G72-86, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23104557

ABSTRACT

Creosote bush-derived nordihydroguaiaretic acid (NDGA), a lipoxygenase inhibitor, possesses antioxidant properties and functions as a potent antihyperlipidemic agent in rodent models. Here, we examined the effect of chronic NDGA treatment of ob/ob mice on plasma dyslipidemia, hepatic steatosis, and changes in hepatic gene expression. Feeding ob/ob mice a chow diet supplemented with either low (0.83 g/kg diet) or high-dose (2.5 g/kg diet) NDGA for 16 wk significantly improved plasma triglyceride (TG), inflammatory chemokine levels, hyperinsulinemia, insulin sensitivity, and glucose intolerance. NDGA treatment caused a marked reduction in liver weight and TG content, while enhancing rates of fatty acid oxidation. Microarray analysis of hepatic gene expression demonstrated that NDGA treatment altered genes for lipid metabolism, with genes involved in fatty acid catabolism most significantly increased. NDGA upregulated the mRNA and nuclear protein levels of peroxisome proliferator-activated receptor α (PPARα), and the activated (phosphorylated) form of AMP-activated kinase. NDGA increased PPARα promoter activity in AML12 hepatocytes and also prevented the fatty acid suppression of PPARα expression. In contrast, PPARα siRNA abrogated the stimulatory effect of NDGA on fatty acid catabolism. Likewise, no stimulatory effect of NDGA on hepatic fatty acid oxidation was observed in the livers of PPARα-deficient mice, but the ability of NDGA to reverse fatty liver conditions was unaffected. In conclusion, the beneficial actions of NDGA on dyslipidemia and hepatic steatosis in ob/ob mice are exerted primarily through enhanced fatty acid oxidation via PPARα-dependent pathways. However, PPARα-independent pathways also contribute to NDGA's action to ameliorate hepatic steatosis.


Subject(s)
Hypolipidemic Agents/therapeutic use , Lipid Metabolism Disorders/drug therapy , Lipoxygenase Inhibitors/therapeutic use , Liver/metabolism , Masoprocol/therapeutic use , PPAR alpha/physiology , Adipokines/metabolism , Animals , Diet , Endoplasmic Reticulum Stress/physiology , Endoribonucleases/metabolism , Fatty Acids/metabolism , Fatty Liver/drug therapy , Glucose Tolerance Test , Leptin/deficiency , Lipid Metabolism/genetics , Lipoproteins, VLDL/metabolism , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Multigene Family , Protein Serine-Threonine Kinases/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Triglycerides/biosynthesis
8.
Pain ; 154(3): 350-360, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23218523

ABSTRACT

Fatty acid ethanolamides (FAEs), which include palmitoylethanolamide (PEA) and oleoylethanolamide (OEA), are endogenous agonists of peroxisome proliferator-activated receptor-α (PPAR-α) and important regulators of the inflammatory response. They are degraded in macrophages by the lysosomal cysteine amidase, N-acylethanolamine acid amidase (NAAA). Previous studies have shown that pharmacological inhibition of NAAA activity suppresses macrophage activation in vitro and causes marked anti-inflammatory effects in vivo, which is suggestive of a role for NAAA in the control of inflammation. It is still unknown, however, whether NAAA-mediated FAE deactivation might regulate pain signaling. The present study examined the effects of ARN077, a potent and selective NAAA inhibitor recently disclosed by our group, in rodent models of hyperalgesia and allodynia caused by inflammation or nerve damage. Topical administration of ARN077 attenuated, in a dose-dependent manner, heat hyperalgesia and mechanical allodynia elicited in mice by carrageenan injection or sciatic nerve ligation. The antinociceptive effects of ARN077 were prevented by the selective PPAR-α antagonist GW6471 and did not occur in PPAR-α-deficient mice. Furthermore, topical ARN077 reversed the allodynia caused by ultraviolet B radiation in rats, and this effect was blocked by pretreatment with GW6471. Sciatic nerve ligation or application of the proinflammatory phorbol ester 12-O-tetradecanoylphorbol 13-acetate decreased FAE levels in sciatic nerve and skin tissue, respectively. ARN077 reversed these biochemical effects. The results identify ARN077 as a potent inhibitor of intracellular NAAA activity, which is active in vivo by topical administration. The findings further suggest that NAAA regulates peripheral pain initiation by interrupting endogenous FAE signaling at PPAR-α.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Analgesics/therapeutic use , Carbamates/therapeutic use , Endocannabinoids/physiology , Enzyme Inhibitors/therapeutic use , Ethers, Cyclic/therapeutic use , Hyperalgesia/drug therapy , Oleic Acids/physiology , PPAR alpha/physiology , Pain Perception/drug effects , Amides , Amidohydrolases/genetics , Amidohydrolases/physiology , Analgesics/administration & dosage , Analgesics/pharmacology , Animals , Burns/drug therapy , Burns/etiology , Carbamates/administration & dosage , Carbamates/pharmacology , Carrageenan/toxicity , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Ethanolamines , Ethers, Cyclic/administration & dosage , Ethers, Cyclic/pharmacology , HEK293 Cells , Humans , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Lysosomes/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR alpha/agonists , PPAR alpha/deficiency , Pain Perception/physiology , Palmitic Acids , Radiation Injuries/drug therapy , Radiation Injuries/etiology , Rats , Recombinant Fusion Proteins/physiology , Sciatic Nerve/injuries , Tetradecanoylphorbol Acetate/toxicity , Ultraviolet Rays/adverse effects
9.
Am J Physiol Endocrinol Metab ; 301(5): E1022-32, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21862726

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) control energy homeostasis. In this study, we showed that farnesol, a naturally occurring ligand of PPARs, could ameliorate metabolic diseases. Obese KK-Ay mice fed a high-fat diet (HFD) containing 0.5% farnesol showed significantly decreased serum glucose level, glucosuria incidence, and hepatic triglyceride contents. Farnesol-containing HFD upregulated the mRNA expressions of PPARα target genes involved in fatty acid oxidation in the liver. On the other hand, farnesol was not effective in upregulating the mRNA expressions of PPARγ target genes in white adipose tissues. Experiments using PPARα-deficient [(-/-)] mice revealed that the upregulation of fatty acid oxidation-related genes required PPARα function, but the suppression of hepatic triglyceride accumulation was partially PPARα-dependent. In hepatocytes isolated from the wild-type and PPARα (-/-) mice, farnesol suppressed triglyceride synthesis. In luciferase assay, farnesol activated both PPARα and the farnesoid X receptor (FXR) at similar concentrations. Moreover, farnesol increased the mRNA expression level of a small heterodimer partner known as one of the FXR target genes and decreased those of sterol regulatory element-binding protein-1c and fatty acid synthase in both the wild-type and PPARα (-/-) hepatocytes. These findings suggest that farnesol could improve metabolic abnormalities in mice via both PPARα-dependent and -independent pathways and that the activation of FXR by farnesol might contribute partially to the PPARα-independent hepatic triglyceride content-lowering effect. To our knowledge, this is the first study on the effect of the dual activators of PPARα and FXR on obesity-induced metabolic disorders.


Subject(s)
Farnesol/pharmacology , Farnesol/therapeutic use , Metabolic Diseases/drug therapy , Metabolic Diseases/prevention & control , PPAR alpha/physiology , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/prevention & control , Diet, High-Fat , Hepatocytes/drug effects , Hepatocytes/metabolism , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Male , Metabolic Diseases/genetics , Mice , Mice, Knockout , Obesity/etiology , Obesity/genetics , Obesity/prevention & control , PPAR alpha/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology , Terpenes/pharmacology , Terpenes/therapeutic use , Triglycerides/metabolism
10.
J Neuroendocrinol ; 23(7): 591-600, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21554431

ABSTRACT

Palmitoylethanolamide (PEA) regulates many pathophysiological processes in the central nervous system, including pain perception, convulsions and neurotoxicity, and increasing evidence points to its neuroprotective action. In the present study, we report that PEA, acting as a ligand of peroxisome-proliferator activated receptor (PPAR)-α, might regulate neurosteroidogenesis in astrocytes, which, similar to other glial cells and neurones, have the enzymatic machinery for neurosteroid de novo synthesis. Accordingly, we used the C6 glioma cell line and primary murine astrocytes. In the mitochondrial fraction from cells stimulated with PEA, we demonstrated an increase in steroidogenic acute regulatory protein (StAR) and cytochrome P450 enzyme (P450scc) expression, both comprising proteins considered to be involved in crucial steps of neurosteroid formation. The effects of PEA were completely blunted by GW6471, a selective PPAR-α antagonist, or by PPAR-α silencing by RNA interference. Accordingly, allopregnanolone (ALLO) levels were increased in supernatant of PEA-treated astrocytes, as revealed by gas chromatography-mass spectrometry, and this effect was inhibited by GW6471. Moreover, PEA showed a protective effect, reducing malondialdehyde formation in cells treated with l-buthionine-(S,R)-sulfoximine, a glutathione depletor and, interestingly, the effect of PEA was partially inhibited by finasteride, a 5α-reductase inhibitor. A similar profile of activity was demonstrated by ALLO and the lack of an additive effect with PEA suggests that the reduction of oxidative stress by PEA is mediated through ALLO synthesis. The present study provides evidence indicating the involvement of the saturated acylethanolamide PEA in ALLO synthesis through PPAR-α in astrocytes and explores the antioxidative activity of this molecule, confirming its homeostatic and protective role both under physiological and pathological conditions.


Subject(s)
Astrocytes/metabolism , Brain Neoplasms/metabolism , Glioma/metabolism , PPAR alpha/physiology , Palmitic Acids/pharmacology , Pregnanolone/biosynthesis , Amides , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/drug effects , Brain Neoplasms/pathology , Cell Culture Techniques , Cell Line, Tumor , Cells, Cultured , Drug Evaluation, Preclinical , Endocannabinoids , Ethanolamines , Glioma/pathology , Mice , Mice, Inbred BALB C , PPAR alpha/genetics , PPAR alpha/metabolism , Rats , Up-Regulation/drug effects
11.
Mol Nutr Food Res ; 53(9): 1143-55, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19655312

ABSTRACT

Hop (Humulus lupulus L.) is an essential ingredient of beer, where it provides the typical bitter taste, but is also applied in traditional folk medicine for sedative and antibacterial purposes. In this study, we demonstrate and compare the anti-inflammatory effect of various classes of hop bitter acids (HBA), including alpha-acids (AA), beta-acids (BA), and iso-alpha-acids (IAA), in fibroblasts, which are important players in the inflammatory response. All three studied classes of HBA blocked the tumor necrosis factor alpha (TNF)-induced production of the cytokine IL6, and inhibited the transactivation of the pro-inflammatory transcription factors nuclear factor kappa B (NF-kappaB), activator protein-1 (AP-1), and cAMP-response element-binding protein (CREB). In this respect, the six-membered ring compounds AA and BA showed equal potency, whereas the five-membered ring compounds, IAA, were effective only when used at higher concentrations. Furthermore, with regard to the mechanism of NF-kappaB suppression, we excluded a possible role for glucocorticoid receptor alpha (GRalpha), peroxisome proliferators-activated receptor alpha/gamma (PPARalpha or PPARgamma), nuclear receptors (NRs) that are also known to inhibit inflammation by directly interfering with the activity of pro-inflammatory transcription factors. Interestingly, combining hop acids and selective agonists for GRalpha, PPARalpha, or PPARgamma resulted in additive inhibition of NF-kappaB activity after TNF treatment, which may open up new avenues for combinatorial anti-inflammatory strategies with fewer side effects. Finally, systemic administration of HBA efficiently inhibited acute local inflammation in vivo.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Humulus/chemistry , PPAR alpha/physiology , PPAR gamma/physiology , Receptors, Glucocorticoid/physiology , Animals , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/physiology , Female , Humans , Interleukin-6/biosynthesis , Mice , Mice, Inbred C57BL , NF-kappa B/antagonists & inhibitors , Transcription Factor AP-1/physiology , Transcription, Genetic/drug effects
12.
J Ethnopharmacol ; 124(3): 544-50, 2009 Jul 30.
Article in English | MEDLINE | ID: mdl-19454308

ABSTRACT

AIM: The present study is designed to investigate the effect of shanzha (Crataegus pinnatifida) on obesity or dyslipidemia induced by high-fat diet in hamsters and characterize the role of PPARalpha in this action of shanzha. MATERIALS AND METHODS: We induced dyslipidemia and obesity in hamsters using high-fat diet and treated hamsters with shanzha or vehicle for 7 days. We measured the body weight, adipose tissue weights, and food intake of hamsters. Plasma total cholesterol (TC), triglyceride (TG), LDL-cholesterol (LDL-C) and HDL-cholesterol (HDL-C) were determined at the beginning and end of this treatment. Effect of shanzha on adipogenesis was examined in vitro and change of PPARalpha was analyzed using Western blot. RESULTS: The food intake, body weights, and weights of both brown and white adipose tissues were markedly reduced in hamsters receiving shanzha as compared with the vehicle-treated control. Plasma levels of TC, TG and LDL-C were decreased by this shanzha treatment while HDL-C was elevated. The effects of shanzha were reversed by the combined treatment with PPARalpha antagonist, MK886. Shanzha inhibited the fat droplet accumulation in 3T3-L1 adipocytes in a dose-dependent manner and this effect was abolished by MK886. Western blot results showed activation of PPARalpha by shanzha in hamster adipose tissue. CONCLUSION: We suggest that shanzha could activate PPARalpha to improve dyslipidemia or obesity.


Subject(s)
Anti-Obesity Agents/therapeutic use , Dietary Fats/pharmacology , Drugs, Chinese Herbal/pharmacology , Hypolipidemic Agents/therapeutic use , 3T3 Cells , Adipocytes/drug effects , Adiposity/drug effects , Animals , Azo Compounds , Blotting, Western , Body Weight/drug effects , Cell Differentiation/drug effects , Coloring Agents , Cricetinae , Diet , Dyslipidemias/blood , Dyslipidemias/drug therapy , Eating/drug effects , Indicators and Reagents , Male , Mesocricetus , Mice , PPAR alpha/physiology
13.
Diabetes ; 58(7): 1532-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19401423

ABSTRACT

OBJECTIVE: Fibroblast growth factor (FGF)-21 improves insulin sensitivity and lipid metabolism in obese or diabetic animal models, while human studies revealed increased FGF-21 levels in obesity and type 2 diabetes. Given that FGF-21 has been suggested to be a peroxisome proliferator-activator receptor (PPAR) alpha-dependent regulator of fasting metabolism, we hypothesized that free fatty acids (FFAs), natural agonists of PPARalpha, might modify FGF-21 levels. RESEARCH DESIGN AND METHODS: The effect of fatty acids on FGF-21 was investigated in vitro in HepG2 cells. Within a randomized controlled trial, the effects of elevated FFAs were studied in 21 healthy subjects (13 women and 8 men). Within a clinical trial including 17 individuals, the effect of insulin was analyzed using an hyperinsulinemic-euglycemic clamp and the effect of PPARgamma activation was studied subsequently in a rosiglitazone treatment trial over 8 weeks. RESULTS: Oleate and linoleate increased FGF-21 expression and secretion in a PPARalpha-dependent fashion, as demonstrated by small-interfering RNA-induced PPARalpha knockdown, while palmitate had no effect. In vivo, lipid infusion induced an increase of circulating FGF-21 in humans, and a strong correlation between the change in FGF-21 levels and the change in FFAs was observed. An artificial hyperinsulinemia, which was induced to delineate the potential interaction between elevated FFAs and hyperinsulinemia, revealed that hyperinsulinemia also increased FGF-21 levels in vivo, while rosiglitazone treatment had no effect. CONCLUSIONS: The results presented here offer a mechanism explaining the induction of the metabolic regulator FGF-21 in the fasting situation but also in type 2 diabetes and obesity.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Fasting/physiology , Fatty Acids, Nonesterified/metabolism , Fibroblast Growth Factors/metabolism , PPAR gamma/physiology , Thiazolidinediones/therapeutic use , Cell Line , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Female , Fibroblast Growth Factors/genetics , Gene Expression Regulation/drug effects , Glucose Clamp Technique , Glycerol/pharmacology , Homeostasis , Humans , Hyperinsulinism/metabolism , Hyperinsulinism/physiopathology , Hypoglycemic Agents/therapeutic use , Lecithins/pharmacology , Male , Obesity/complications , Obesity/metabolism , PPAR alpha/genetics , PPAR alpha/physiology , PPAR gamma/genetics , RNA, Messenger/genetics , Reference Values , Rosiglitazone
14.
Subcell Biochem ; 49: 3-47, 2008.
Article in English | MEDLINE | ID: mdl-18751906

ABSTRACT

The liver is a major site of fatty acid synthesis and degradation. Transcriptional regulation is one of several mechanisms controlling hepatic metabolism of fatty acids. Two transcription factors, namely SREBP1-c and PPARalpha, appear to be the main players controlling synthesis and degradation of fatty acids respectively. This chapter briefly presents fatty acid metabolism. The first part focuses on SREBP1-c contribution to the control of gene expression relevant to fatty acid synthesis and the main mechanisms of activation for this transcriptional program. The second part reviews the evidence for the involvement of PPARalpha in the control of fatty acid degradation and the key features of this nuclear receptor. Finally, the third part aims at summarizing recent advances in our current understanding of how these two transcription factors fit in the regulatory networks that sense hormones or nutrients, including cellular fatty acids, and govern the transcription of genes implicated in hepatic fatty acid metabolism.


Subject(s)
Fatty Acids/metabolism , Gene Expression Regulation , Liver/metabolism , PPAR alpha/physiology , Sterol Regulatory Element Binding Protein 1/physiology , Acetyl-CoA Carboxylase/metabolism , Acetyltransferases/metabolism , Adipose Tissue/metabolism , Animals , Dietary Fats, Unsaturated/pharmacology , Eating , Fatty Acid Elongases , Fatty Acid Synthases/metabolism , Fatty Acids/biosynthesis , Glucose/metabolism , Homeostasis , Humans , Hydroxylation , Insulin/physiology , Linoleoyl-CoA Desaturase/metabolism , Metabolic Networks and Pathways , Mitochondria, Liver/metabolism , Oxidation-Reduction , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stearoyl-CoA Desaturase/metabolism
15.
J Med Food ; 11(2): 252-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18598166

ABSTRACT

Elevated plasma lipids, glucose, insulin, and fatty liver are among components of metabolic syndrome, a phenotypic pattern that typically precedes the development of Type 2 diabetes. Animal studies show that intake of anthocyanins reduces hyperlipidemia, obesity, and atherosclerosis and that anthocyanin-rich extracts may exert these effects in association with altered activity of tissue peroxisome proliferator-activated receptors (PPARs). However, studies are lacking to test this correlation using physiologically relevant, whole food sources of anthocyanins. Tart cherries are a rich source of anthocyanins, and whole cherry fruit intake may also affect hyperlipidemia and/or affect tissue PPARs. This hypothesis was tested in the Dahl Salt-Sensitive rat having insulin resistance and hyperlipidemia. For 90 days, Dahl rats were pair-fed AIN-76a-based diets supplemented with either 1% (wt:wt) freeze-dried whole tart cherry or with 0.85% additional carbohydrate to match macronutrient and calorie provision. After 90 days, the cherry-enriched diet was associated with reduced fasting blood glucose, hyperlipidemia, hyperinsulinemia, and reduced fatty liver. The cherry diet was also associated with significantly enhanced hepatic PPAR-alpha mRNA, enhanced hepatic PPAR-alpha target acyl-coenzyme A oxidase mRNA and activity, and increased plasma antioxidant capacity. In conclusion, physiologically relevant tart cherry consumption reduced several phenotypic risk factors that are associated with risk for metabolic syndrome and Type 2 diabetes. Tart cherries may represent a whole food research model of the health effects of anthocyanin-rich foods and may possess nutraceutical value against risk factors for metabolic syndrome and its clinical sequelae.


Subject(s)
Anthocyanins/administration & dosage , Fatty Liver/drug therapy , Hyperlipidemias/drug therapy , PPAR alpha/genetics , Phytotherapy , Prunus/chemistry , Animals , Diabetes Mellitus, Type 2/prevention & control , Diet , Fruit/chemistry , Insulin Resistance , Liver/chemistry , Male , Metabolic Syndrome/prevention & control , PPAR alpha/physiology , RNA, Messenger/analysis , Rats , Rats, Inbred Dahl
16.
Eur J Nutr ; 47(2): 80-6, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18320257

ABSTRACT

BACKGROUND: alpha-Amino-beta-carboxymuconate-epsilon-semialdehyde decarboxylase (ACMSD) is a key enzyme in NAD biosynthesis from tryptophan. Dietary polyunsaturated fatty acids (PUFA) have been shown to suppress hepatic ACMSD activity and its mRNA level in rat. However the mechanism of the suppressive action has not been clarified yet. Although the phenomena that fatty acids suppress the expression of ACMSD in rat liver have been established in vivo experiment, it is still obscure whether the effect of fatty acids on the expression of the enzyme is caused by its direct or indirect action, because there have been very few investigations performed in vitro. AIM OF THE STUDY: In this study, to examine whether down-regulation of ACMSD mRNA by PUFA involves peroxisome proliferator-activated receptor (PPAR) alpha mediated mechanism or not, we investigated the effect of PUFA on the ACMSD expression by using primary cultured rat hepatocytes. METHODS: For this purpose we investigated the effect of PUFA (linoleic acid and eicosapentanoic acid) on the ACMSD mRNA level in primary-cultured rat hepatocytes and compared its effect with that of WY-14,643 (a PPARalpha agonist). After the incubation of hepatocytes with fatty acids, WY-14,643 and/or MK886 (a PPARalpha antagonist), mRNA levels of ACMSD and a peroxisome marker enzyme acyl-CoA oxidase (ACO) were determined by competitive reverse transcription-polymerase chain reaction (RT-PCR) method. RESULTS: ACMSD mRNA level in primary hepatocytes were decreased by the incubation with high concentrations of linoleic acid, eicosapentaenoic acid (EPA) and WY-14,643. The appearance of ACO mRNA by WY-14,643 was remarkably increased, and those by linoleic acid and EPA were increased less than that by WY-14,643. Moreover, the suppression of ACMSD mRNA and the augmentation of ACO mRNA by WY-14,643 were inhibited by MK886, but the suppression by PUFA was not substantially affected by MK886. CONCLUSIONS: The present study suggesting that the mechanism of decrease in ACMSD mRNA level by PUFA was different from that by WY-14,643, and that there would be any pathway other than PPARalpha mediated one for PUFA to regulate ACMSD expression.


Subject(s)
Carboxy-Lyases/biosynthesis , Dietary Fats, Unsaturated/pharmacology , Fatty Acids, Unsaturated/pharmacology , Hepatocytes/enzymology , PPAR alpha/physiology , Animals , Carboxy-Lyases/genetics , Cells, Cultured , Eicosapentaenoic Acid/pharmacology , Enzyme Repression , Indoles/pharmacology , Linoleic Acid/pharmacology , PPAR alpha/antagonists & inhibitors , Pyrimidines/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
17.
Int J Mol Med ; 21(4): 439-45, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18360689

ABSTRACT

Perfluorinated compounds (PFCs) have been employed as surface treatment agents in a variety of products. Perfluorooctanoic acid (PFOA), a PFC that is found globally in the environment and in human tissues, has been increasing significantly in serum levels over the past 50 years. Here, we demonstrated that PFOA inhibits feeding behavior as potently as the endogenous peroxisome proliferator-activated receptor (PPAR)-alpha ligand, oleoylethanolamide (OEA), via the activation of PPAR-alpha, the vagal nerve and hypothalamic neuropeptides. Peripherally administered PFOA decreased food intake as potently as OEA. PFOA decreased gastric emptying and increased the expression level of the gene encoding urocortin 1 in the hypothalamus and the immunoreaction for urocortin 1 in the paraventricular nucleus. Vagotomy attenuated the inhibitory effects of PFOA on feeding. The inhibition of food intake and body-weight gain by PFOA was completely mitigated in PPAR-alpha-/-mice. Our studies demonstrated that the ubiquitous environmental pollutant PFOA works as an imitator of OEA mimicking its action in the feeding regulatory system, providing a new mode of action as represented by environmental 'anorexigens'.


Subject(s)
Caprylates/toxicity , Environmental Pollutants/toxicity , Feeding Behavior/drug effects , Feeding Behavior/physiology , Fluorocarbons/toxicity , PPAR alpha/physiology , Animals , Base Sequence , Caprylates/administration & dosage , DNA Primers/genetics , Eating/drug effects , Eating/genetics , Eating/physiology , Environmental Pollutants/administration & dosage , Female , Fluorocarbons/administration & dosage , Food Deprivation , Gastric Emptying/drug effects , Humans , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Mice , Mice, Knockout , PPAR alpha/deficiency , PPAR alpha/genetics , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Urocortins/genetics , Vagotomy
18.
PLoS One ; 3(2): e1681, 2008 Feb 27.
Article in English | MEDLINE | ID: mdl-18301758

ABSTRACT

BACKGROUND: The effect of dietary fats on human health and disease are likely mediated by changes in gene expression. Several transcription factors have been shown to respond to fatty acids, including SREBP-1c, NF-kappaB, RXRs, LXRs, FXR, HNF4alpha, and PPARs. However, it is unclear to what extent these transcription factors play a role in gene regulation by dietary fatty acids in vivo. METHODOLOGY/PRINCIPAL FINDINGS: Here, we take advantage of a unique experimental design using synthetic triglycerides composed of one single fatty acid in combination with gene expression profiling to examine the effects of various individual dietary fatty acids on hepatic gene expression in mice. We observed that the number of significantly changed genes and the fold-induction of genes increased with increasing fatty acid chain length and degree of unsaturation. Importantly, almost every single gene regulated by dietary unsaturated fatty acids remained unaltered in mice lacking PPARalpha. In addition, the majority of genes regulated by unsaturated fatty acids, especially docosahexaenoic acid, were also regulated by the specific PPARalpha agonist WY14643. Excellent agreement was found between the effects of unsaturated fatty acids on mouse liver versus cultured rat hepatoma cells. Interestingly, using Nuclear Receptor PamChip(R) Arrays, fatty acid- and WY14643-induced interactions between PPARalpha and coregulators were found to be highly similar, although several PPARalpha-coactivator interactions specific for WY14643 were identified. CONCLUSIONS/SIGNIFICANCE: We conclude that the effects of dietary unsaturated fatty acids on hepatic gene expression are almost entirely mediated by PPARalpha and mimic those of synthetic PPARalpha agonists in terms of regulation of target genes and molecular mechanism. Use of synthetic dietary triglycerides may provide a novel paradigm for nutrigenomics research.


Subject(s)
Dietary Fats/pharmacology , Gene Expression Regulation/drug effects , PPAR alpha/physiology , Triglycerides/pharmacology , Animals , Cells, Cultured , Dietary Fats/chemical synthesis , Gene Expression Profiling , Liver/metabolism , Rats , Structure-Activity Relationship , Transcription Factors , Triglycerides/chemical synthesis
19.
Semin Nephrol ; 27(6): 609-20, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18061843

ABSTRACT

Although proteomics studies the global expression of proteins, metabolomics characterizes and quantifies their end products: the metabolites, produced by an organism under a certain set of conditions. From this perspective it is apparent that proteomics and metabolomics are complementary and when joined allow a fuller appreciation of an organism's phenotype. Our studies using (1)H-nuclear magnetic resonance spectroscopic analysis showed the presence of glucose, amino acids, and trichloroacetic acid cycle metabolites in the urine after 48 hours of cisplatin administration. These metabolic alterations precede changes in serum creatinine. Biochemical studies confirmed the presence of glucosuria, but also showed the accumulation of nonesterified fatty acids, and triglycerides in serum, urine, and kidney tissue, despite increased levels of plasma insulin. These metabolic alterations were ameliorated by the use of fibrates. We propose that the injury-induced metabolic profile may be used as a biomarker of cisplatin-induced nephrotoxicity. These studies serve to illustrate that metabolomic studies add insight into pathophysiology not provided by proteomic analysis alone.


Subject(s)
Acute Kidney Injury/metabolism , Proteins/analysis , Proteomics/methods , Acute Kidney Injury/chemically induced , Cisplatin/toxicity , Fatty Acids/analysis , Glucose/metabolism , Humans , Hyperglycemia/chemically induced , Hyperglycemia/prevention & control , Insulin/blood , Kidney/metabolism , Magnetic Resonance Spectroscopy , PPAR alpha/physiology , Triglycerides/metabolism
20.
Eur J Pharmacol ; 565(1-3): 158-65, 2007 Jun 22.
Article in English | MEDLINE | ID: mdl-17434475

ABSTRACT

Vascular cell adhesion molecule-1 (VCAM-1) is involved in several diseases, including chronic inflammation and atherosclerosis. Inhibition of the expression of this adhesion molecule is one of the key targets of anti-inflammatory, anti-cancer and anti-atherosclerotic drugs. Gynostemma pentaphyllum is a traditional medicine widely used in the treatment of respiratory inflammation, hyperlipidemia and atherosclerosis. However, its molecular mechanisms of action are still largely unknown. Gypenoside XLIX, a dammarane-type glycoside, is a prominent component of G. pentaphyllum. We have recently demonstrated Gypenoside XLIX to be a selective peroxisome proliferator-activated receptor (PPAR)-alpha activator. Here we demonstrate that Gypenoside XLIX concentration-dependently (0-300 microM) inhibited VCAM-1 promoter activity after induction by cytokine tumor necrosis factor (TNF)-alpha in human umbilical vein endothelial cells (HUVECs) transfected with promoter-reporter construct pVCAM-1-LUC. Furthermore, Gypenoside XLIX inhibited TNF-alpha-induced VCAM-1 mRNA and protein overexpression in HUVECs. The result of the enzyme immunoassay demonstrated that Gypenoside XLIX inhibited TNF-alpha-induced increase in cell surface VCAM-1 protein levels in HUVECs. In the present study we show that activities of Gypenoside XLIX are similar to those of Wy-14643, a potent synthetic PPAR-alpha activator. Furthermore, Gypenoside XLIX-induced inhibition on TNF-alpha-stimulated VCAM-1 promoter hyperactivity was completely abolished by a selective blocker of PPAR-alpha, MK-886. Thus, our findings suggest that Gypenoside XLIX inhibits cytokine-induced VCAM-1 overexpression and hyperactivity in human endothelial cells via a PPAR-alpha-dependent pathway. These data provide new insight into the rational basis of the use of the traditional Chinese herbal medicine G. pentaphyllum in the treatment of inflammatory and cardiovascular diseases, including atherosclerosis.


Subject(s)
Endothelial Cells/drug effects , PPAR alpha/drug effects , Saponins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/genetics , Cells, Cultured , Endothelial Cells/metabolism , Gene Expression Regulation/drug effects , Humans , PPAR alpha/physiology , Promoter Regions, Genetic , Vascular Cell Adhesion Molecule-1/analysis
SELECTION OF CITATIONS
SEARCH DETAIL