Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Nutrients ; 13(11)2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34836021

ABSTRACT

Marine n-3 fatty acids are well known to have health benefits. Recently, krill oil, which contains phospholipids, has been in the spotlight as an n-3 PUFA-containing oil. Euphausia pacifica (E. pacifica), also called North Pacific krill, is a small, red crustacean similar to shrimp that flourishes in the North Pacific Ocean. E. pacifica oil contains 8-hydroxyeicosapentaenoic acid (8-HEPE) at a level more than 10 times higher than Euphausia superba oil. 8-HEPE can activate the transcription of peroxisome proliferator-activated receptor alpha (PPARα), PPARγ, and PPARδ to levels 10, 5, and 3 times greater than eicosapentaenoic acid, respectively. 8-HEPE has beneficial effects against metabolic syndrome (reduction in body weight gain, visceral fat area, amount of gonadal white adipose tissue, and gonadal adipocyte cell size), dyslipidemia (reduction in serum triacylglycerol and low-density lipoprotein cholesterol and induction of serum high-density lipoprotein cholesterol), atherosclerosis, and nonalcoholic fatty liver disease (reduction in triglyceride accumulation and hepatic steatosis in the liver) in mice. Further studies should focus on the beneficial effects of North Pacific krill oil products and 8-HEPE on human health.


Subject(s)
Eicosapentaenoic Acid/analogs & derivatives , Euphausiacea/chemistry , Fish Oils/pharmacology , Animals , Atherosclerosis/blood , Atherosclerosis/therapy , Dyslipidemias/blood , Dyslipidemias/therapy , Humans , Liver/metabolism , Metabolic Syndrome/blood , Metabolic Syndrome/therapy , Mice , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/therapy , PPAR alpha/blood , PPAR delta/blood , PPAR gamma/blood
2.
Hepatol Commun ; 5(12): 2035-2051, 2021 12.
Article in English | MEDLINE | ID: mdl-34558841

ABSTRACT

Accumulation of cytotoxic bile acids (BAs) during cholestasis can result in liver failure. Glucuronidation, a phase II metabolism pathway responsible for BA detoxification, is regulated by peroxisome proliferator-activated receptor alpha (PPARα). This study investigates the efficacy of adjunct fenofibrate therapy to up-regulate BA-glucuronidation and reduce serum BA toxicity during cholestasis. Adult patients with primary biliary cholangitis (PBC, n = 32) and primary sclerosing cholangitis (PSC, n = 23), who experienced an incomplete response while receiving ursodiol monotherapy (13-15 mg/kg/day), defined as serum alkaline phosphatase (ALP) ≥ 1.5 times the upper limit of normal, received additional fenofibrate (145-160 mg/day) as standard of care. Serum BA and BA-glucuronide concentrations were measured by liquid chromatography-mass spectrometry. Combination therapy with fenofibrate significantly decreased elevated serum ALP (-76%, P < 0.001), aspartate transaminase, alanine aminotransferase, bilirubin, total serum BAs (-54%), and increased serum BA-glucuronides (+2.1-fold, P < 0.01) versus ursodiol monotherapy. The major serum BA-glucuronides that were favorably altered following adjunct fenofibrate include hyodeoxycholic acid-6G (+3.7-fold, P < 0.01), hyocholic acid-6G (+2.6-fold, P < 0.05), chenodeoxycholic acid (CDCA)-3G (-36%), and lithocholic acid (LCA)-3G (-42%) versus ursodiol monotherapy. Fenofibrate also up-regulated the expression of uridine 5'-diphospho-glucuronosyltransferases and multidrug resistance-associated protein 3 messenger RNA in primary human hepatocytes. Pearson's correlation coefficients identified strong associations between serum ALP and metabolic ratios of CDCA-3G (r2  = 0.62, P < 0.0001), deoxycholic acid (DCA)-3G (r2  = 0.48, P < 0.0001), and LCA-3G (r2  = 0.40, P < 0.001), in ursodiol monotherapy versus control. Receiver operating characteristic analysis identified serum BA-glucuronides as measures of response to therapy. Conclusion: Fenofibrate favorably alters major serum BA-glucuronides, which correlate with reduced serum ALP levels and improved outcomes. A PPARα-mediated anti-cholestatic mechanism is involved in detoxifying serum BAs in patients with PBC and PSC who have an incomplete response on ursodiol monotherapy and receive adjunct fenofibrate. Serum BA-glucuronides may serve as a noninvasive measure of treatment response in PBC and PSC.


Subject(s)
Bile Acids and Salts/metabolism , Cholangitis, Sclerosing/drug therapy , Cholestasis/drug therapy , Fenofibrate/administration & dosage , Glucuronides/blood , Liver Cirrhosis, Biliary/drug therapy , Adult , Cholangitis, Sclerosing/blood , Cholestasis/blood , Drug Therapy, Combination , Female , Hepatocytes/metabolism , Humans , Liver/metabolism , Liver Cirrhosis, Biliary/blood , Liver Function Tests , Male , Middle Aged , PPAR alpha/blood , Retrospective Studies , Treatment Outcome , Up-Regulation/drug effects , Ursodeoxycholic Acid/administration & dosage , Young Adult
3.
Toxicol Sci ; 168(2): 519-534, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30657992

ABSTRACT

Epidemiological studies have reported positive associations between serum perfluorooctanoic acid (PFOA) and total and non-high-density lipoprotein cholesterol (non-HDL-C) although the magnitude of effect of PFOA on cholesterol lacks consistency. The objectives of this study were to evaluate the effect of PFOA on plasma cholesterol and triglyceride metabolism at various plasma PFOA concentrations relevant to humans, and to elucidate the mechanisms using APOE*3-Leiden.CETP mice, a model with a human-like lipoprotein metabolism. APOE*3-Leiden.CETP mice were fed a Western-type diet with PFOA (10, 300, 30 000 ng/g/d) for 4-6 weeks. PFOA exposure did not alter plasma lipids in the 10 and 300 ng/g/d dietary PFOA dose groups. At 30 000 ng/g/d, PFOA decreased plasma triglycerides (TG), total cholesterol (TC), and non-HDL-C, whereas HDL-C was increased. The plasma lipid alterations could be explained by decreased very low-density lipoprotein (VLDL) production and increased VLDL clearance by the liver through increased lipoprotein lipase activity. The concomitant increase in HDL-C was mediated by decreased cholesteryl ester transfer activity and changes in gene expression of proteins involved in HDL metabolism. Hepatic gene expression and pathway analysis confirmed the changes in lipoprotein metabolism that were mediated for a major part through activation of the peroxisome proliferator-activated receptor (PPAR)α. Our data confirmed the findings from a phase 1 clinical trial in humans that demonstrated high serum or plasma PFOA levels resulted in lower cholesterol levels. The study findings do not show an increase in cholesterol at environmental or occupational levels of PFOA exposure, thereby indicating these findings are associative rather than causal.


Subject(s)
Caprylates/toxicity , Fluorocarbons/toxicity , Lipoproteins/blood , Triglycerides/blood , Water Pollutants, Chemical/toxicity , Animals , Apolipoprotein E3/genetics , Caprylates/blood , Cholesterol, HDL/blood , Cholesterol, VLDL/blood , Dose-Response Relationship, Drug , Fluorocarbons/blood , Humans , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice, Transgenic , PPAR alpha/blood , Water Pollutants, Chemical/blood
4.
J Lipid Res ; 59(11): 2140-2152, 2018 11.
Article in English | MEDLINE | ID: mdl-30158201

ABSTRACT

PPARα (PPARA), expressed in most oxidative tissues, is a major regulator of lipid homeostasis; hepatic PPARA plays a critical role during the adaptive fasting response by promoting FA oxidation (FAO). To clarify whether extrahepatic PPARA activity can protect against lipid overload when hepatic PPARA is impaired, lipid accumulation was compared in WT (Ppara+/+), total body Ppara-null (Ppara-/-), and hepatocyte-specific Ppara-null (PparaΔHep) mice that were fasted for 24 h. Histologic staining indicated reduced lipid accumulation in PparaΔHep versus Ppara-/- mice, and biochemical analyses revealed diminished medium- and long-chain FA accumulation in PparaΔHep mouse livers. Hepatic PPARA target genes were suppressed in both mouse models. Serum FFAs increased in all genotypes after fasting but were highest in Ppara-/- mice. In PparaΔHep mice, FAO genes were increased in brown adipose tissue, heart, and muscle, and total lipase activity was elevated in the muscle and heart, suggesting increased lipid utilization. Thus, extrahepatic PPARA activity reduces systemic lipid load when hepatic lipid metabolism is impaired by elevating FAO and lipase activity in other tissues and, as a result, protects against fasting-induced hepatosteatosis. This has important clinical implications in disease states with impaired hepatic PPARA function, such as nonalcoholic steatohepatitis and nonalcoholic fatty liver disease.


Subject(s)
Liver/metabolism , PPAR alpha/metabolism , Animals , Fasting/blood , Gas Chromatography-Mass Spectrometry , Lipid Metabolism/physiology , Male , Malondialdehyde/blood , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/blood , Oxidation-Reduction , PPAR alpha/blood , PPAR alpha/genetics
5.
Diabetes Metab Syndr ; 12(5): 711-714, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29699951

ABSTRACT

BACKGROUND: Metabolic syndrome (MS) is considered one of the major worldwide epidemics. It accounts for billions of cardiovascular disease events and deaths. Till now, major basics of MS are not fully clarified. Peroxisome Proliferator-Activated Receptor-α (PPARα) displays a ligand-activated transcription factor. It is involved in the regulation of many metabolic processes including inflammation, lipid, and glucose metabolism. Therefore, this study investigated the leucocytic expression of PPARα in a metabolic patient in comparison to healthy controls. METHODS: 100 subjects with MS were recruited, in addition to 100 subjects without any obvious metabolic disorders as healthy controls. Expression of PPARα and CD 36 were analyzed on different leucocytic populations using optimized flow-cytometric analysis. Correlations of the expression of both indexes with different clinical and laboratory parameters were analyzed. RESULTS: The eosinophilic expression of PPARα was found to be lower in subjects with MS in comparison to the healthy controls (p value 0.001). Also, PPARα expression, on most of the leucocytic populations, was inversely correlated with waist circumferences among the study populations. CONCLUSION: Circulated eosinophilic expression of PPARα protein is reduced in MS subjects. This conclusion may explain the endothelial dysfunction and obesity associated with MS, as well as it may help in the management of this worldwide health problem.


Subject(s)
Eosinophils/metabolism , Leukocytes, Mononuclear/metabolism , Metabolic Syndrome/blood , Metabolic Syndrome/diagnosis , PPAR alpha/biosynthesis , PPAR alpha/blood , Adult , Case-Control Studies , Egypt/epidemiology , Female , Gene Expression Regulation , Humans , Male , Metabolic Syndrome/epidemiology , Middle Aged , PPAR alpha/genetics
6.
J Nutr ; 148(3): 379-388, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29546309

ABSTRACT

Background: High-fat meals induce postprandial inflammation. Resveratrol is a polyphenol known to prevent comorbidities associated with cardiovascular disease and exerts an anti-inflammatory action. There is also an increasing body of evidence supporting the role of curcumin, a polyphenol from the curcuminoid family, as a modulator of proinflammatory processes. Objective: The objectives of this study were to investigate the following: 1) the bioavailability of resveratrol consumed in combination with curcumin after consumption of a high-fat meal; and 2) the acute combined effects of this combination on the postprandial inflammatory response of subjects with abdominal obesity. Methods: In a double blind, crossover, randomized, placebo-controlled study, 11 men and 11 postmenopausal women [mean ± SD age: 62 ± 5 y; mean ± SD body mass index (in kg/m2): 29 ± 3] underwent a 6-h oral fat tolerance test on 2 occasions separated by 1-2 wk: once after consumption of a dietary supplement (200 mg resveratrol and 100 mg curcumin, Res/Cur) and once after consumption of a placebo (cellulose). Plasma concentrations of total resveratrol and its major metabolites as well as inflammatory markers, adhesion molecules, and whole blood NFκB1 and PPARA gene expression were measured during both fat tolerance tests. Results: Kinetics of resveratrol and identified metabolites revealed rapid absorption patterns but also relatively limited bioavailability based on free resveratrol concentrations. Supplementation with Res/Cur did not modify postprandial variations in circulating inflammatory markers (C-reactive protein, IL-6, IL-8, monocyte chemoattractant protein-1) and adhesion molecules [soluble E-selectin, soluble vascular cell adhesion molecule-1 (sVCAM-1), soluble intercellular adhesion molecule-1] compared to placebo (PTreatment×Time > 0.05). However, Res/Cur significantly decreased the cumulative postprandial response of sVCAM-1, compared to placebo (incremental area under the curve -4643%, P = 0.01). Postprandial variations of whole-blood PPARA and NFKB1 gene expression were not different between Res/Cur and placebo treatments. Conclusions: Acute supplementation with Res/Cur has no impact on the postprandial inflammation response to a high-fat meal in abdominally obese older adults. Further studies are warranted to examine how resveratrol and curcumin may alter the vascular response to a high-fat meal. This trial was registered at clinicaltrials.gov as NCT01964846.


Subject(s)
Curcumin/pharmacology , Dietary Fats/adverse effects , Dietary Supplements , Inflammation Mediators/blood , Inflammation/etiology , Obesity, Abdominal/complications , Resveratrol/pharmacology , Aged , Anti-Inflammatory Agents/pharmacology , Area Under Curve , Biological Availability , C-Reactive Protein/metabolism , Chemokine CCL2/blood , Cross-Over Studies , Curcumin/metabolism , Dietary Fats/administration & dosage , Double-Blind Method , Drug Combinations , Female , Humans , Inflammation/blood , Interleukins/blood , Male , Middle Aged , PPAR alpha/blood , Plant Extracts/pharmacology , Postprandial Period , Resveratrol/metabolism
7.
Mol Med Rep ; 16(1): 49-56, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28498479

ABSTRACT

Although peroxisome proliferator-activated receptor (PPAR)-α has been reported to be involved in preventing acute lung injury (ALI), the molecular regulation of post­ALI lung recovery remains to be fully elucidated. The aim of the present study was to characterize the mechanism by which PPAR­α prevents ALI and examine the role of PPAR­α in the recovery of lung function following acute respiratory distress syndrome (ARDS). Reverse transcription­quantitative­polymerase chain reaction and western blot analyses suggested that PPAR­α was effective in suppressing transforming growth factor (TGF)­ß1 in HLF cells and RAW 264.7 cells. In an ALI mouse model, PPAR­α treatment prior to stimulation with lipopolysaccharide (LPS) resulted in a decrease in the expression of TGF­ß1 in bronchoalveolar lavage fluid (BALF), peripheral blood and splenocytes. The injection of a virus expressing short hairpin PPAR­α into mice following LPS treatment resulted in a dose­dependent increase in lung resistance index and decrease in dynamic compliance, and a significant increase in BALF protein, which indicated PPAR­α was essential for the recovery of lung function following ALI. Of note, the serum expression of PPAR­α was inversely correlated with TGF­ß1 and negatively correlated with disease severity in patients with ARDS. These data suggested that PPAR­α was essential for the recovery of lung function following ALI by the suppression of TGF­ß1, which reveals a previously unappreciated mechanism controlling post­ALI lung recovery.


Subject(s)
PPAR alpha/metabolism , Recovery of Function , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/physiopathology , Transforming Growth Factor beta1/metabolism , Aged , Animals , Cell Line , Disease Models, Animal , Female , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Humans , Lipopolysaccharides/adverse effects , Macrophages/metabolism , Male , Mice , Middle Aged , PPAR alpha/blood , PPAR alpha/genetics , PPAR alpha/pharmacology , Respiratory Distress Syndrome/etiology , Respiratory Function Tests , Transforming Growth Factor beta1/blood , Transforming Growth Factor beta1/genetics
8.
Med Sci Monit ; 23: 1363-1370, 2017 Mar 19.
Article in English | MEDLINE | ID: mdl-28315901

ABSTRACT

BACKGROUND In clinics, patients with type 2 diabetes complicated with non-alcoholic fatty liver disease (NAFLD) have been shown to receive significant improvements in blood glucose levels, lipid levels, and liver function after sitagliptin treatment, although the mechanism of drug action remains poorly understood. This study investigated the possible mechanism of sitagliptin on lipid metabolism of NAFLD mice. MATERIAL AND METHODS Male C57/BL6 mice were induced for NAFLD via 16 weeks of a high-fat diet, and were treated with 15 mg/kg/day sitagliptin for 16 consecutive weeks. Blood lipid levels were measured and samples were stained with hematoxylin and eosin (H&E) and oil red staining for liver pathology and lipid deposition. Serum levels of fibroblast growth factor (FGF)-9 and FGF-21 were quantified by enzyme-linked immunosorbent assay (ELISA). Peroxisome proliferator-activated receptor (PPAR)-α, and cAMP reactive element binding homolog (CREBH) were measured by Western blotting, while fatty acid synthase and carnitine palmitoyltransferase 1 (CPT1) mRNA levels were assayed by RT-PCR. RESULTS Compared to the control group, the NAFLD model mice had liver fatty disease, lower serum FGF-21 and FGF-19 levels, elevated serum lipid levels, depressed PPAR-α, CREBH, and CPT1 expression, and enhanced FAS expression (p<0.05). Sitagliptin treatment depressed blood lipid levels, increased serum FGF-21 and FGF-19 levels, PPAR-α, CREBH, and CPT1 expression, and suppressed FAS expression (p<0.05). CONCLUSIONS Sitagliptin can protect liver tissue and modulate lipid metabolism in NAFLD mice via elevating FGF-21 and FGF-19, upregulating liver PPAR-a and CREBH levels, and mediating expression levels of key enzymes for lipid metabolism.


Subject(s)
Lipid Metabolism/drug effects , Sitagliptin Phosphate/metabolism , Sitagliptin Phosphate/therapeutic use , Animals , Carnitine O-Palmitoyltransferase/blood , Carnitine O-Palmitoyltransferase/metabolism , Cyclic AMP Response Element-Binding Protein/blood , Cyclic AMP Response Element-Binding Protein/metabolism , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat , Disease Models, Animal , Fatty Liver/drug therapy , Fatty Liver/metabolism , Fibroblast Growth Factors/drug effects , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , PPAR alpha/blood , PPAR alpha/metabolism , Sitagliptin Phosphate/pharmacology
9.
Med Sci Monit ; 21: 735-9, 2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25758678

ABSTRACT

BACKGROUND: The aim of this study was to observe the change in plasma PPARs (peroxisome proliferator-activated receptors) level during various periods and in different subtypes in migraine patients. MATERIAL AND METHODS: We divided 227 patients with migraine into 2 main groups: the attack period group (n=98) and the attack-free period group (n=129). Patients were further divided into 4 subgroups according to whether they had aura symptoms. The control group consisted of 100 healthy subjects. We collected the clinical data of patients and measured the plasma levels of PPARs using enzyme-linked immunoassay (ELISA). We used SPSS software for statistical analysis. RESULTS: We found no significant difference in age, BMI, blood pressure, or blood lipid level among migraine patients during the headache attack period and during the headache-free period compared with the control group. The PPARα and PPARß/δ levels during the headache attack period were significantly higher than during the headache free period and in healthy controls. The PPARγ levels during the headache attack period were significantly lower than those during the headache-free period and in the healthy control group. The PPARs levels during the headache attack period were significantly different from those during the headache-free period, regardless of presence or absence of aura. The PPARs levels during the headache-free period were not significantly different from those of the healthy control group. The level of PPARs has no significant differences between migraine with aura group and without aura group, regardless of whether headache attack. CONCLUSIONS: PPARs involved in the pathogenesis of migraine. Presence of absence of aura had no obvious effect on PPARs level.


Subject(s)
Migraine Disorders/blood , Peroxisome Proliferator-Activated Receptors/blood , Adult , Female , Humans , Male , PPAR alpha/blood , PPAR gamma/blood , PPAR-beta/blood
10.
J Lipid Res ; 56(2): 254-65, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25510248

ABSTRACT

PPARα is well known as a master regulator of lipid metabolism. PPARα activation enhances fatty acid oxidation and decreases the levels of circulating and cellular lipids in obese diabetic patients. Although PPARα target genes are widely known, little is known about the alteration of plasma and liver metabolites during PPARα activation. Here, we report that metabolome analysis-implicated upregulation of many plasma lysoGP species during bezafibrate (PPARα agonist) treatment. In particular, 1-palmitoyl lysophosphatidylcholine [LPC(16:0)] is increased by bezafibrate treatment in both plasma and liver. In mouse primary hepatocytes, the secretion of LPC(16:0) increased on PPARα activation, and this effect was attenuated by PPARα antagonist treatment. We demonstrated that Pla2g7 gene expression levels in the murine hepatocytes were increased by PPARα activation, and the secretion of LPC(16:0) was suppressed by Pla2g7 siRNA treatment. Interestingly, LPC(16:0) activates PPARα and induces the expression of PPARα target genes in hepatocytes. Furthermore, we showed that LPC(16:0) has the ability to recover glucose uptake in adipocytes induced insulin resistance. These results reveal that LPC(16:0) is induced by PPARα activation in hepatocytes; LPC(16:0) contributes to the upregulation of PPARα target genes in hepatocytes and the recovery of glucose uptake in insulin-resistant adipocytes.


Subject(s)
Lysophosphatidylcholines/blood , Lysophosphatidylcholines/metabolism , Metabolomics , PPAR alpha/blood , PPAR alpha/metabolism , 3T3-L1 Cells , Animals , Bezafibrate/pharmacology , Chromatography, High Pressure Liquid , Hepatocytes/drug effects , Hepatocytes/metabolism , Insulin Resistance , Lipid Metabolism/drug effects , Male , Mice , RNA, Small Interfering
11.
Pediatr Obes ; 9(6): 463-70, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24277691

ABSTRACT

WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT: The expression of specific genes in peripheral blood cells (PBCs) may be used as biomarkers of the metabolic status. High levels of expression of CPT1A, SLC27A2, INSR, LEPR, FASN and PPARα in PBCs are indicative of a lower risk for the insulin resistant or dyslipidaemic state associated with obesity in children. Breastfeeding seems to confer protective effects against obesity and its related metabolic problems. WHAT THIS STUDY ADDS: Children who had been breastfed showed higher expression levels of SLC27A2, FASN, PPARα and INSR in PBCs compared with formula-fed subjects. The relationship of the PBC transcript levels of SLC27A2, INSR, FASN and PPARα with insulin resistance and dyslipidaemia may be dependent on the type of infant feeding (breast vs. formula). The transcript levels of the mentioned biomarkers could be useful to distinguish the formula-fed children who are at higher risk of metabolic alterations. BACKGROUND: Blood-cell transcripts have showed to be good biomarkers of metabolic alterations and their use in early detection and prevention of future disorders is promising. OBJECTIVE: This study aimed to examine the relation between previously proposed transcriptional biomarkers of metabolic health (SLC27A2, CPT1A, FASN, PPARα, INSR, LEPR) in peripheral blood cells and the type of infant feeding in a subset of children from the IDEFICS (Identification and Prevention of Dietary- and Lifestyle-Induced Health Effects in Children and Infants) cohort. SUBJECTS: A total of 237 children aged 2-9 years from eight European countries were studied. RESULTS: Breastfed children showed higher expression levels of SLC27A2, FASN, PPARα and INSR, and lower risk of being overweight and of having high plasma triglyceride levels vs. formula-fed children. Besides, overweight formula-fed children presented higher HOMA-index than overweight breastfed children (1.90 vs. 1.62); however, this negative effect was absent in formula-fed children with high expression of SLC27A2. Moreover, formula-fed children with low expression of SLC27A2, FASN, PPARα and INSR presented higher triglyceride levels than subjects with high expression of these genes (77.7 mg dL(-1) vs. 44.8 mg dL(-1) ). This difference was absent in breastfed children. CONCLUSIONS: Protective effects of breastfeeding are reflected in higher expression levels of SLC27A2, FASN, PPARα and INSR in blood cells. These biomarkers may also serve to discriminate the formula-fed children that are at higher risk of metabolic alterations.


Subject(s)
Antigens, CD/blood , Breast Feeding , Coenzyme A Ligases/blood , Fatty Acid Synthase, Type I/blood , PPAR alpha/blood , Pediatric Obesity/blood , Receptor, Insulin/blood , Biomarkers/blood , Body Mass Index , Child , Child, Preschool , Europe/epidemiology , Female , Gene Expression , Humans , Infant , Infant Nutritional Physiological Phenomena , Infant, Newborn , Insulin Resistance , Male , Pediatric Obesity/epidemiology , Pediatric Obesity/prevention & control , Pregnancy , Risk Factors
12.
Mar Environ Res ; 92: 43-51, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24025588

ABSTRACT

Harbour seals as top predators and indicators for ecosystem health are exposed to increasing pressure caused by anthropogenic activities in their marine environment. After their lactation period of about 24 days pups are weaned and left to hunt on their own. Little is known about the development of their immune system and a better understanding of anthropogenic impacts on the general health and immune system of harbour seal pups is needed. mRNA transcription of six immuno-relevant biomarkers was analysed in 13 abandoned harbour seal pups from the North Sea, fostered at the Seal Centre Friedrichskoog, Germany. RNAlater blood samples were taken at admission, day 22 and before release and analysed using RT-qPCR. Significant differences in HSP70, cytokine IL-2 and xenobiotic biomarkers AHR, ARNT and PPARα transcription were found between admission, during rehabilitation and before release. Highest levels at admission may result from dehydration, handling, transport and contaminant exposure via lactation. The significant decrease is linked to health improvement, feeding and adaptation. The increase before release is suspected to be due to infection pressure and contaminant exposure from feeding on fish. Molecular biomarkers are a sensitive tool to evaluate health and pollutant exposure and useful to serve as early warning indicators, monitoring and case-by-case tool for marine mammals in human care and the wild.


Subject(s)
Biomarkers/blood , Immune System/physiology , Phoca/blood , Phoca/immunology , RNA, Messenger/analysis , Water Pollutants, Chemical/toxicity , Xenobiotics/pharmacokinetics , Age Factors , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/blood , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Body Weight , Cytokines/blood , Cytokines/genetics , Female , HSP72 Heat-Shock Proteins/blood , HSP72 Heat-Shock Proteins/genetics , Hematocrit , Male , North Sea , PPAR alpha/blood , PPAR alpha/genetics , Receptors, Aryl Hydrocarbon/blood , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Xenobiotics/toxicity
13.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 44(3): 383-7, 2013 May.
Article in Chinese | MEDLINE | ID: mdl-23898518

ABSTRACT

OBJECTIVE: To determine the mRNA expressions of PPARalpha and PPARbeta in peripheral blood mononuclear cells of non-vavular hypertensive atrial fibrillation (AF) patients and elucidate its possible role in the pathogenesis of AF. METHODS: Peripheral blood samples were collected from 103 patients with hypertensive AF (persistent AF: 55, paroxysmal AF: 48) and 50 age-adjusted hypertension patients without AF. The mRNA expressions of PPARalpha, PPARbeta, interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-alpha) in monocytes were detected by using a Real time polymerase chain reaction. The concentrations of high sensitive C-reactive protein (CRP) and interleukin-1 (IL-1) were measured by immunoenzymetric method. RESULTS: The PPARalpha mRNA expression level was persistently decreased in hypertensive non-AF group, paroxysmal AF group, and persistent AF group (1.34 +/- 0.17, 1.09 +/- 0.23, 0.85 +/- 0.22), while the difference was statistically significant (P < 0.001; respectively). TNF-alpha mRNA, IL-6 mRNA,CRP and IL-1 persistently increased in hypertensive non-AF group, paroxysmal AF group, persistent AF group, also the difference was statistically significant (P < 0. 001; respectively). The difference of PPARbeta mRNA was not statistically significant between non-AF group, paroxysmal AF group and persistent AF group. Left atrial diameter (LAD) was in positive correlation with CRP, IL-1, IL-6 mRNA and TNF-alpha mRNA (P < 0.05). PPARalpha mRNA level was in negative correlation with CRP, IL-1, IL-6 mRNA and TNF-alpha mRNA, the correlation coefficient was -0.519, -0.532, -0.491 and -0.528, respectively (P < 0.05). CONCLUSION: In hypertensive patients with AF, increased inflammatory cytokines were associated with atrial remodeling and lead to the development of atrial fibrillation; PPARalpha was negatively correlated with these inflammatory cytokines and may play a vital role in the process of atrial fibrillation development.


Subject(s)
Atrial Fibrillation/blood , Hypertension/complications , Leukocytes, Mononuclear/metabolism , PPAR alpha/blood , PPAR-beta/blood , Aged , Atrial Fibrillation/etiology , C-Reactive Protein/genetics , C-Reactive Protein/metabolism , Female , Humans , Hypertension/blood , Interleukin-6/genetics , Interleukin-6/metabolism , Male , Middle Aged , PPAR alpha/genetics , PPAR-beta/genetics , RNA, Messenger/blood , RNA, Messenger/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
14.
Toxicology ; 309: 52-60, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23619606

ABSTRACT

Maternal exposure to di(2-ethylhexyl) phthalate (DEHP) decreased the plasma triglyceride in prepartum mice. To identify the fatty acid (FA) species involved and to understand the underlying mechanisms, pregnant Sv/129 wild-type (mPPARα), peroxisome proliferator-activated receptor α-null (Pparα-null) and humanized PPARα (hPPARα) mice were treated with diets containing 0%, 0.01%, 0.05% or 0.1% DEHP. Dams were dissected on gestational day 18 together with fetuses, and on postnatal day 2 together with newborns. n-3/n-6 polyunsaturated, saturated, and monounsaturated FAs in maternal plasma and in liver of wild-type offspring, and representative enzymes for FA desaturation and elongation in maternal liver, were measured. The plasma levels of linoleic acid, α-linolenic acid, palmitic acid and oleic acid were higher in the pregnant control mPPARa mice than in Ppara-null and hPPARa mice. DEHP exposure significantly decreased the levels of these four FAs only in pregnant mPPARα mice. Plasma levels of many FAs were higher in pregnant mice than in postpartum ones in a genotype-independent manner, while it was lower in the livers of fetuses than pups. DEHP exposure slightly increased hepatic arachidonic acid, α-linolenic acid, palmitoleic acid and oleic acid in fetuses, but not in pups. However, DEHP exposure did not clearly influence FA desaturase 1 and 2 nor elongase 2 and 5 expressions in the liver of all maternal mice. Taken together, the levels of plasma four FAs with shorter carbon chains were higher in pregnant mPPARα mice than in other genotypes, and DEHP exposure decreased these specific FA concentrations only in mPPARα mice, similarly to triglyceride levels.


Subject(s)
Diethylhexyl Phthalate/toxicity , Fatty Acids/blood , Maternal Exposure/adverse effects , Paternal Exposure/adverse effects , Animals , Animals, Newborn , Biomarkers/blood , Female , Humans , Male , Mice , Mice, 129 Strain , Mice, Knockout , Mice, Transgenic , PPAR alpha/blood , PPAR alpha/deficiency , Pregnancy
15.
Am J Physiol Endocrinol Metab ; 303(10): E1212-21, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22990076

ABSTRACT

PPARδ is a transcription factor regulating the expression of genes involved in oxidative metabolism, which may regulate blood cholesterols through transcription of oxidative and lipoprotein metabolism genes. To determine the association of skeletal muscle PPARδ content with blood lipids and lipoproteins before and following exercise, overweight and obese men (n = 9) and women (n = 7) were recruited; age, BMI, body fat percentage, and Vo(2max) were (means ± SE) 45 ± 2.5 yr, 31.9 ± 1.4 kg/m(-2), 41.1 ± 1.5%, and 26.0 ± 1.3 mLO(2)·kg(-1)·min(-1), respectively. Subjects performed 12 wk of endurance exercise training (3 sessions/wk, progressing to 500 kcal/session). To assess the acute exercise response, subjects performed a single exercise session on a treadmill (70% Vo(2max), 400 kcal energy expenditure) before and after training. Muscle and blood samples were obtained prior to any exercise and 24 h after each acute exercise session. Muscle was analyzed for protein content of PPARδ, PPARα, PGC-1α, AMPKα, and the oxidative and lipoprotein markers FAT/CD36, CPT I, COX-IV, LPL, F(1) ATPase, ABCAI, and LDL receptor. Blood was assessed for lipids and lipoproteins. Repeated-measures ANOVA revealed no influence of sex on measured outcomes. PPARδ, PGC-1α, FAT/CD36, and LPL content were enhanced following acute exercise, whereas PPARα, AMPKα, CPT I, and COX-IV content were enhanced only after exercise training. PPARδ content negatively correlated with total and LDL cholesterol concentrations primarily in the untrained condition (r ≤ -0.4946, P < 0.05), whereas AMPKα was positively correlated with HDL cholesterol concentrations regardless of exercise (r ≥ 0.5543, P < 0.05). Our findings demonstrate exercise-induced expression of skeletal muscle PPARs and their target proteins, and this expression is associated with improved blood lipids and lipoproteins in obese adults.


Subject(s)
Adenylate Kinase/metabolism , Exercise/physiology , Lipids/blood , Lipoproteins/blood , Muscle, Skeletal/metabolism , Obesity/metabolism , PPAR delta/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/blood , Adenylate Kinase/blood , Biopsy , Blotting, Western , CD36 Antigens/blood , Cholesterol/blood , Cohort Studies , Energy Metabolism/physiology , Female , Heat-Shock Proteins/blood , Humans , Lipoprotein Lipase/blood , Male , Middle Aged , Muscle, Skeletal/enzymology , Obesity/blood , Obesity/enzymology , PPAR alpha/blood , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Proton-Translocating ATPases/blood , Receptors, LDL/blood , Statistics, Nonparametric , Transcription Factors/blood
16.
J Asian Nat Prod Res ; 14(7): 678-87, 2012.
Article in English | MEDLINE | ID: mdl-22582804

ABSTRACT

Sinomenine (SN, 1) is a pure compound extracted from the Sinomenium acutum plant. We investigated the protective effects and mechanism of action of SN in a rat model of doxorubicin (DOX)-induced nephrosis. Nephrosis was induced by a single dose of 5 mg/kg DOX, and DOX-treated rats received a daily i.p. injection of 10 or 30 mg/kg SN, or saline (n = 6). Urine and serum biochemical parameters, serum TNF-α and IL-1ß levels, nephrin, podocin, α-actinin-4, and peroxisome proliferator-activated receptor-α (PPAR-α) protein expression, and renal ultrastructure were examined at day 28. Compound 1 significantly attenuated the effect of DOX on urine and serum biochemical parameters. Electron microscopy demonstrated that 1 suppressed DOX-induced increases in foot process width. Compared with those in control rats, nephrin, podocin, and PPAR-α protein expressions decreased in the glomeruli of DOX-treated rats, and this effect was significantly attenuated by 1. However, no appreciable alterations were observed in the expression level of α-actinin-4. DOX significantly increased serum TNF-α and IL-1ß compared with those in control rats, and 1 significantly reduced the serum levels of TNF-α and IL-1ß. SN ameliorates DOX-induced nephrotic syndrome in rats, resulting in a modulation of renal nephrin, podocin expression, and thereby protecting podocytes from injury.


Subject(s)
Doxorubicin/adverse effects , Doxorubicin/pharmacology , Morphinans/pharmacology , Nephrosis/chemically induced , Animals , Doxorubicin/analysis , Interleukin-1beta/analysis , Interleukin-1beta/blood , Interleukin-1beta/urine , Intracellular Signaling Peptides and Proteins/analysis , Intracellular Signaling Peptides and Proteins/blood , Intracellular Signaling Peptides and Proteins/urine , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Kidney Glomerulus/ultrastructure , Male , Membrane Proteins/analysis , Membrane Proteins/blood , Membrane Proteins/urine , Models, Biological , Molecular Structure , Morphinans/therapeutic use , PPAR alpha/analysis , PPAR alpha/blood , PPAR alpha/urine , Rats , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/urine
17.
J Med Food ; 15(5): 495-503, 2012 May.
Article in English | MEDLINE | ID: mdl-22551412

ABSTRACT

The protective effect of raspberry ketone against nonalcoholic steatohepatitis (NASH) was tested by using a high-fat diet-induced NASH model, and its mechanism was explored. Forty Sprague-Dawley rats with a 1:1 male to female ratio were randomly divided into five groups: the normal control (NC) group (n=8) fed normal diet for 8 weeks, the model control (MC) group (n=8) fed high-fat diet (82% standard diet, 8.3% yolk powder, 9.0% lard, 0.5% cholesterol, and 0.2% sodium taurocholate), and the raspberry ketone low-dose (0.5%) (RKL) group (n=8), the raspberry ketone middle-dose (1%) (RKM) group (n=8), and the raspberry ketone high-dose (2%) (RKH) group (n=8) fed high-fat diet for 4 weeks. After 8 weeks of experiment, all the rats were sacrificed, and blood lipid parameters (total cholesterol [TC], triglycerides [TG], high-density lipoprotein cholesterol [HDL-C], and low-density lipoprotein cholesterol [LDL-C]), liver function parameters (serum alanine aminotransferase [ALT], aspartate aminotransferase [AST], and alkaline phosphatase [ALP]), leptin (LEP), free fatty acid (FFA), tumor necrosis factor α (TNF-α), blood glucose (GLU), and insulin (INS) with calculated INS resistance index (IRI) and INS-sensitive index (ISI) were measured in rats. Therefore, we determined the peroxisome proliferator-activated receptor (PPAR)-α activity in liver homogenate and the levels of low-density lipoprotein receptor (LDLR), high-sensitivity C-reactive protein (hs-CRP), adiponection (APN), superoxide dismutase, and malondialdehyde (MDA). The liver tissues of rats in each group were imaged by electron microscopy with hematoxylin-eosin as the staining agent. The levels of TG, TC, LDL-C, ALT, AST, ALP, GLU, INS, IRI, FFA, LEP, TNF-α, MDA, and hs-CRP of MC rats were significantly increased (P<.05, P<.01). Therefore, the levels of HDL-C, ISI, PPAR-α, LDLR, and APN were significantly decreased (P<.05, P<.01). Compared with the MC group, each parameter in the RKL, RKM, and RKH groups was significantly improved (P<.05, P<.01). Thus raspberry ketone was an effective intervention for NASH in rats. It was believed that raspberry ketone had a dual effect of liver protection and fat reduction, and the mechanism was probably mediated by alleviation of fatty degeneration of liver cells, decreased liver inflammation, correction of dyslipidemia, reversal of LEP and INS resistance, and improved antioxidant capacity.


Subject(s)
Butanones/therapeutic use , Fatty Liver/prevention & control , Lipids/blood , Liver/drug effects , Phytotherapy , Plant Extracts/therapeutic use , Rosaceae/chemistry , Adipokines/blood , Animals , Biomarkers/blood , Blood Glucose/metabolism , Butanones/pharmacology , C-Reactive Protein/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Fatty Liver/blood , Fatty Liver/etiology , Female , Fruit/chemistry , Insulin/blood , Insulin Resistance , Liver/metabolism , Male , Non-alcoholic Fatty Liver Disease , PPAR alpha/blood , Plant Extracts/pharmacology , Rats , Rats, Sprague-Dawley , Transaminases/blood , Tumor Necrosis Factor-alpha/blood
18.
Environ Health Prev Med ; 17(6): 444-56, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22407906

ABSTRACT

OBJECTIVES: The aim of this study was to identify the molecular mechanisms underlying high-fat and high-cholesterol (HFC) diet-induced steatohepatitis and associated liver fibrosis progression in a novel stroke-prone, spontaneously hypertensive 5/Dmcr (SHRSP5/Dmcr) rat model. METHODS: SHRSP5/Dmcr rats were given the control or HFC-diet for 2, 8, and 16 weeks. Plasma and hepatic gene expression of key molecules involved in fatty acid oxidation, inflammation, oxidative stress, and fibrosis were subsequently analyzed. RESULTS: Rats fed the HFC-diet showed increased plasma tumor necrosis factor-α (TNF-α) and hepatic p50/p65 signals, but reduced hepatic Cu(2+)/Zn(2+)-superoxide dismutase across the treatment period and reduced plasma total adiponectin at 8 weeks. In HFC-diet-fed rats, transforming growth factor-ß1 (TGF-ß1) was elevated prior to the appearance of obvious liver fibrosis pathology at 2 weeks, followed by elevations in platelet-derived growth factor-B (PDGF-B) and α-smooth muscle actin (α-SMA), corresponding to evident liver fibrosis, at 8 weeks and by α(1) type I collagen production at 16 weeks. The HFC-diet increased hepatic total cholesterol accumulation, although hepatic triglyceride declined by 0.3-fold from 2 to 16 weeks due to reduced hepatic triglyceride synthesis, as suggested by the diacylglycerol acyltransferase 1 and 2 measurements. CONCLUSIONS: TNF-α and p50/p65 molecular signals appeared to be major factors for HFC-diet-induced hepatic inflammation and oxidative stress facilitating liver disease progression. While the up-regulation of TGF-ß1 prior to the appearance of any evident liver fibrosis could be an early signal for progressive liver fibrosis, elevated PDGF-B and α-SMA levels signified evident liver fibrosis at 8 weeks, and subsequent increased α(1) type I collagen production and reduced triglyceride synthesis indicated extensive liver fibrosis at 16 weeks in this novel SHRSP5/Dmcr model.


Subject(s)
Cholesterol, Dietary/adverse effects , Diet, High-Fat/adverse effects , Disease Models, Animal , Fatty Liver/etiology , Liver Cirrhosis/etiology , Rats , Animals , Biomarkers/blood , Blotting, Western , Cholesterol/blood , Dietary Fats/blood , Disease Progression , Enzyme-Linked Immunosorbent Assay , Fatty Liver/blood , Fatty Liver/pathology , Intercellular Signaling Peptides and Proteins/blood , Liver/pathology , Liver Cirrhosis/blood , Liver Cirrhosis/pathology , Male , PPAR alpha/blood , RNA, Messenger/metabolism , Rats, Inbred SHR , Real-Time Polymerase Chain Reaction
19.
J Sports Med Phys Fitness ; 52(1): 102-6, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22327093

ABSTRACT

AIM: The present study aimed at investigating the effect of a 3-week training on biomarkers in professional soccer players during the preseason preparation-period. METHODS: Eight participants (age 22.5±2.2 yrs) were enrolled in the study. During the physical preparation period players have attended a training program (51.9 hours) formulated by coaches of "Equipe-Sicilia-2009". RESULTS: At rest, the lipid profile, the creatine kinase (CK), the lactic-acid dehydrogenase (LDH) and the expression of nuclear receptors peroxisome-proliferator-activated receptors (PPAR α/γ) were analyzed before starting and after 3 weeks of training. The plasma level of CK in our samples showed great variability already in the baseline: value was on average nearly 500 IU/l showed that a large amount of these athletes were a high responders. This biomarker showed a reduction (P<0.01) after 3 weeks of training. No modifications were found in the LDH plasma level, in the lipid profile and in the expression of mRNA of PPAR α/γ and also no significant person's correlations were found among variables. CONCLUSION: In conclusion, we retain that those basal biomarkers, except CK, are not able to assist coaches to better understand training adaptations and overreaching mechanisms during a 3-week of preseason preparation-period. More studies are necessary to confirm these results.


Subject(s)
Creatine Kinase/blood , L-Lactate Dehydrogenase/blood , PPAR alpha/blood , PPAR gamma/blood , Physical Education and Training , Soccer/physiology , Adult , Biomarkers/blood , Humans , PPAR alpha/genetics , PPAR gamma/genetics , RNA, Messenger/blood , Young Adult
20.
Nutr Metab Cardiovasc Dis ; 22(11): 966-73, 2012 Nov.
Article in English | MEDLINE | ID: mdl-21429719

ABSTRACT

BACKGROUND AND AIMS: To compare the effects of n-3 long chain polyunsaturated fatty acids (n-3 LCPUFA), with those of fenofibrate, on markers of inflammation and vascular function, and on the serum lipoprotein profile in overweight and obese subjects. METHODS AND RESULTS: Twenty overweight and obese subjects participated in a randomized, double-blind, placebo-controlled intervention trial and received 3.7 g/d n-3 fatty acids (providing 1.7 g/d EPA and 1.2 g/d DHA), 200 mg fenofibrate or placebo treatment for 6 weeks separated by a 2 weeks wash-out period. Fish oil and fenofibrate treatment reduced triglyceride (-0.61 ± 0.81 mmol/L, P < 0.001, and -0.34 ± 0.85 mmol/L, P = 0.048, respectively) and increased HDL cholesterol concentrations (0.13 ± 0.21 mmol/L, P = 0.013, and 0.10 ± 0.18 mmol/L, P = 0.076), as reflected by a decrease of large very VLDL particles and increases of large HDL particles and medium size HDL particles. Fish oil increased serum LDL cholesterol concentrations (0.34 ± 0.59 mmol/L, P = 0.013). Fenofibrate reduced concentrations of soluble endothelial selectin (sE-selectin) (-4.1 ± 7.5 ng/mL, P = 0.032), but increased those of macrophage chemoattractant protein 1 (MCP1) (28 ± 55 ng/mL, P = 0.034). Fish oil had no effects on these markers. CONCLUSION: Although n-3 LCPUFA and fenofibrate can both activate PPARα, they have differential effects on cardiovascular risk markers. In overweight and obese subjects fenofibrate (200 mg/d) or n-3 LCPUFA (3.7 g/d) treatment for 6 weeks did not improve markers for low-grade systemic inflammation, while fenofibrate had more profound effects on plasma lipids and markers for vascular activity compared to fish oil. Registration number clinical trials EudraCT 2006-005743-28.


Subject(s)
Biomarkers/analysis , Fatty Acids, Omega-3/pharmacology , Fenofibrate/pharmacology , Inflammation/drug therapy , Obesity/blood , Overweight/blood , Adult , Aged , Biomarkers/blood , Cardiovascular Diseases/prevention & control , Chemokine CCL2/blood , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Docosahexaenoic Acids/administration & dosage , Double-Blind Method , E-Selectin/blood , Female , Fish Oils/administration & dosage , Humans , Male , Metabolic Syndrome/prevention & control , Middle Aged , PPAR alpha/blood , Risk Factors , Triglycerides/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...