Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Dairy Sci ; 103(7): 6557-6568, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32331890

ABSTRACT

Sirtuin 3 (SIRT3), a mitochondrial deacetylase, is a key regulator of energy metabolism in the liver. In nonruminants, the hepatic abundance of SIRT3 is decreased in individuals with nonalcoholic fatty liver diseases, and recovery of SIRT3 alleviates hepatic triacylglycerol (TG) deposition. However, the level of SIRT3 expression and its effects on lipid metabolism in dairy cows have not been characterized. Here we studied the hepatic expression of SIRT3 in cows with fatty liver and the role of SIRT3 in fatty acid metabolism in bovine hepatocytes. This in vivo study involved 10 healthy cows and 10 cows with fatty liver, from which we collected samples of liver tissue and blood. Primary hepatocytes were isolated from Holstein calves and treated with 0, 0.5, or 1.0 mM nonesterified fatty acids (NEFA) for 24 h or transinfected with SIRT3 overexpression adenovirus (Ad-SIRT3)/SIRT3-short interfering (si)RNA for 48 h. Cows with fatty liver displayed lower serum glucose concentrations but higher serum NEFA and ß-hydroxybutyrate concentrations relative to healthy cows. Cows with fatty liver also had significant lower mRNA and protein abundance of hepatic SIRT3. Incubation of primary hepatocytes with NEFA reduced SIRT3 abundance in primary hepatocytes in a dose-dependent manner. Fatty acid (1 mM) treatment also markedly increased the abundance of acetyl-CoA carboxylase 1 (ACC1) and fatty acid synthase (FAS) but significantly decreased the abundance of carnitine palmitoyltransferase I (CPT1A), carnitine palmitoyltransferase II (CPT2), and acyl-CoA oxidase (ACO). Knockdown of SIRT3 by SIRT3-siRNA downregulated the mRNA abundance of CPT1A, CPT2, and ACO. In contrast, overexpression of SIRT3 by Ad-SIRT3 upregulated the mRNA abundance of CPT1A, CPT2, and ACO; downregulated the mRNA abundance of ACC1 and FAS; and consequently, decreased intracellular TG concentrations. Overexpression of SIRT3 ameliorated exogenous NEFA-induced TG accumulation by downregulating the abundance of ACC1 and FAS and upregulating the abundance of CPT1A, CPT2, and ACO in calf hepatocytes. Our data demonstrated that cows with fatty liver had lower hepatic SIRT3 contents, and an increase in SIRT3 abundance by overexpression mitigated TG deposition by modulating the expression of lipid metabolism genes in bovine hepatocytes. These data suggest a possible role of SIRT3 as a therapeutic target for fatty liver disease prevention in periparturient dairy cattle.


Subject(s)
Cattle Diseases/metabolism , Fatty Acids, Nonesterified/administration & dosage , Fatty Liver/veterinary , Lipid Metabolism/drug effects , Sirtuin 3/metabolism , 3-Hydroxybutyric Acid/blood , Acetyl-CoA Carboxylase/drug effects , Acyl-CoA Oxidase/drug effects , Animals , Carnitine O-Palmitoyltransferase/drug effects , Cattle , Cattle Diseases/prevention & control , Fatty Acids/metabolism , Fatty Acids, Nonesterified/blood , Fatty Liver/metabolism , Fatty Liver/prevention & control , Female , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/drug effects , Liver/metabolism , Mitochondria/enzymology , Sirtuin 3/genetics , Triglycerides/metabolism
2.
J Endocrinol ; 219(1): 69-78, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23873539

ABSTRACT

The rat Harderian gland (HG) is an orbital gland producing a copious lipid secretion. Recent studies indicate that its secretory activity is regulated by thyroid hormones. In this study, we found that both isoforms of the thyroid hormone receptor (Trα (Thra) and Trß (Thrb)) are expressed in rat HGs. Although Thra is expressed at a higher level, only Thrb is regulated by triiodothyronine (T3). Because T3 induces an increase in lipid metabolism in rat HGs, we investigated the effects of an animal's thyroid state on the expression levels of carnitine palmitoyltransferase-1A (Cpt1a) and carnitine palmitoyltransferase-1B (Cpt1b) and acyl-CoA oxidase (Acox1) (rate-limiting enzymes in mitochondrial and peroxisomal fatty acid oxidation respectively), as well as on the mitochondrial compartment, thereby correlating mitochondrial activity and biogenesis with morphological analysis. We found that hypothyroidism decreased the expression of Cpt1b and Acox1 mRNA, whereas the administration of T3 to hypothyroid rats increased transcript levels. Respiratory parameters and catalase protein levels provided further evidence that T3 modulates mitochondrial and peroxisomal activities. Furthermore, in hypothyroid rat HGs, the mitochondrial number and their total area decreased with respect to the controls, whereas the average area of the individual mitochondrion did not change. However, the average area of the individual mitochondrion was reduced by ∼50% in hypothyroid T3-treated HGs, and the mitochondrial number and the total area of the mitochondrial compartment increased. The mitochondrial morphometric data correlated well with the molecular results. Indeed, hypothyroid status did not modify the expression of mitochondrial biogenesis genes such as Ppargc1a, Nrf1 and Tfam, whereas T3 treatment increased the expression level of these genes.


Subject(s)
Acyl-CoA Oxidase/metabolism , Carnitine O-Palmitoyltransferase/metabolism , Harderian Gland/metabolism , Hypothyroidism/metabolism , Mitochondrial Turnover/drug effects , Receptors, Thyroid Hormone/metabolism , Triiodothyronine/pharmacology , Acyl-CoA Oxidase/drug effects , Animals , Carnitine O-Palmitoyltransferase/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Harderian Gland/drug effects , Lipid Metabolism/drug effects , Male , Mitochondria/drug effects , Mitochondria/metabolism , Peroxisomes/drug effects , Peroxisomes/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar
3.
J Toxicol Environ Health A ; 71(23): 1526-41, 2008.
Article in English | MEDLINE | ID: mdl-18923995

ABSTRACT

Perfluorooctanesulfonate (PFOS) is one of a class of industrial chemicals known as perfluoroalkyl acids, which have a wide variety of uses as surfactants and stain repellants. The presence of fluorochemical residues in human blood, plasma, or serum from sample populations worldwide is indicative of widespread human exposure. Previous studies demonstrated that PFOS alters fatty acid metabolism in the liver of rodents and that this leads to peroxisome proliferation. This study was undertaken to (1) confirm the effects of PFOS on rat liver, (2) identify additional target organs and systems, and (3) further explore the biochemical and molecular changes associated with PFOS exposure. The results confirmed that liver was a primary target for PFOS. Hepatomegaly, decreased serum triglycerides and cholesterol, and increased expression of the genes for acyl-coenzymeA oxidase 1 (ACOX1) and cytochrome P-450 4A22 (CYP4A22) were indicative of exposure to a peroxisome proliferator. Changes in liver fatty acid profiles included increased total monounsaturated fatty acid levels and decreased total polyunsaturated fatty acids, as well as an increase in linoleic acid levels and a decrease in longer chain fatty acids. These changes were similar to those induced by relatively weak peroxisome proliferators. Disruptions in hepatic fatty acid metabolism may contribute to changes in red blood cell membranes, resulting in increased lysis and cell fragility. Serum thyroid hormone levels were decreased in PFOS-treated rats, while the kidney and cardiovascular systems were not significant targets. Residue analyses indicated that PFOS accumulation in tissues was dose dependent, appearing preferentially in the liver at lower doses but increasing in serum and other organs relative to liver at higher doses.


Subject(s)
Alkanesulfonic Acids/toxicity , Environmental Pollutants/toxicity , Fatty Acids/metabolism , Fluorocarbons/toxicity , Food Contamination , Liver/drug effects , Acyl-CoA Oxidase/drug effects , Animals , Cytochrome P-450 Enzyme System/drug effects , Cytochrome P450 Family 4 , Dose-Response Relationship, Drug , Erythrocyte Deformability/drug effects , Female , Homeostasis , Liver/metabolism , Liver/pathology , Male , Rats
4.
Eur J Nutr ; 46(6): 364-8, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17571209

ABSTRACT

BACKGROUND: The product of the obesity gene (ob), leptin, has a well-recognized role in regulating energy homeostasis. During the period of weight maintenance, circulating leptin concentration reflects total body fat mass. On the other hand, overnutrition is accompanied by progressive hyperleptinemia. In overnourished animals, the elevation in circulating fatty acids results in increased uptake and excessive deposition of lipids within muscle cells. Consequently, triglicerydes overload seems to strongly correlate to the impairment of insulin signaling in skeletal muscle, the primary target for insulin stimulated glucose disposal. High levels of leptin in the course of fat storage may protect non-adipose tissues from lipid accumulation. AIM OF THE STUDY: Here, we aim to evaluate in vitro the relationship between leptin treatment and expression of acyl-CoA oxidase (ACOX), a peroxisomal key enzyme involved in fatty acid catabolism. We also evaluate the adaptive response of cells to a putative oxidative insult, resulting from H(2)O(2) production. METHODS: The effects of increasing levels of leptin, at different times, were assessed on mouse C2C12 myotubes by semiquantitative PCR. Activation pathway was investigated by using extracellular signal-regulated kinase (ERK) and cytosolic phospholipase A(2) (cPLA(2)) inhibitors. Cellular adaptive response to oxidative stress was evaluated by measuring glutathione concentration, oxidized/reduced glutathione ratio and the main antioxidant enzymatic activities. RESULTS: A 1.8-fold increase in ACOX mRNA expression was evident at 20 ng/ml leptin, a dose comparable to that found in hyperleptinemic subjects. The induction was dose-dependent, with an increase of 3-fold at 100 ng/ml; the ability of leptin to stimulate ACOX mRNA reached a maximum at 20 min and was lost in myotubes continuously exposed for more than 1 h. ACOX enzymatic activity followed mRNA changes: it was doubled after 1 h treatment and remained elevated for 24 h. ERK and cPLA(2) pathway is involved, since their inhibitors abrogated the ACOX mRNA induction. Myotubes counteract the resulting oxidative insult by catalase and glutathione peroxidase activation, thus removing H(2)O(2) at the expenses of the reduced glutahione pool. CONCLUSIONS: The present study shows that acute, but not chronic, leptin treatment of C2C12 myotubes induces ACOX expression. Peroxisomal fatty acid oxidation may work together with mitochondrial beta-oxidation to remove excessive lipids from non-adipose tissues, during early stages of overnutrition and before development of leptin resistance.


Subject(s)
Acyl-CoA Oxidase/drug effects , Gene Expression Regulation, Enzymologic , Leptin/pharmacology , Muscle Fibers, Skeletal/enzymology , RNA, Messenger/drug effects , Acyl-CoA Oxidase/metabolism , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Glutathione/metabolism , Humans , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Oxidation-Reduction , Reverse Transcriptase Polymerase Chain Reaction
5.
J Pharmacol Sci ; 104(2): 137-45, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17538229

ABSTRACT

The effects of a peroxisome proliferator activated receptor gamma (PPARgamma) agonist on hepatic stearoyl-CoA desaturase (SCD) in insulin-resistant and obese Zucker fa/fa rats were studied. The administration of pioglitazone, a PPARgamma agonist, to Zucker obese rats greatly improved their insulin sensitivity. The treatment of Zucker obese rats with pioglitazone did not affect the index of fatty acid desaturation of either serum or liver. Hepatic SCD activity and the mRNA level of SCD1 were not changed by treatment of the rats with pioglitazone. The activity of palmitoly-CoA chain elongase, which is involved in the biosynthesis of oleic acid in concert with SCD, was not significantly altered when Zucker obese rats received pioglitazone. Although neither the activity nor mRNA expression of acyl-CoA oxidase was changed by treatment of Zucker obese rats with pioglitazone, the mRNA expressions of both sterol regulatory element-binding protein-1c and acetyl-CoA carboxylase sensitively responded to the challenge by pioglitazone. These results suggest that the insulin sensitivity of insulin-resistant and obese Zucker fa/fa rats is improved by pioglitazone independently of SCD activity.


Subject(s)
Hypoglycemic Agents/pharmacology , Insulin Resistance , PPAR gamma/agonists , Stearoyl-CoA Desaturase/drug effects , Thiazolidinediones/pharmacology , Acyl-CoA Oxidase/drug effects , Acyl-CoA Oxidase/metabolism , Animals , Blood Glucose , Fatty Acids/metabolism , Gene Expression , Glucose Tolerance Test , Insulin/blood , Liver/drug effects , Liver/metabolism , Male , Obesity , Oleic Acid/biosynthesis , Palmitoyl Coenzyme A/metabolism , Pioglitazone , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Zucker , Stearoyl-CoA Desaturase/metabolism
6.
Article in English | MEDLINE | ID: mdl-16099631

ABSTRACT

Both insulin and PPAR-alpha up-modulate hepatic Delta9, Delta6 and Delta5 desaturating enzymes involved in the biosynthesis of mono- and polyunsaturated fatty acids. Currently, we have examined for 9 days the independent and simultaneous effects of daily glargine insulin and fenofibrate administration on the insulinemia, glycemia, hepatic acyl-CoA oxidase activity and mRNAs and enzymatic activities of stearoyl-CoA desaturase-1 (SCD-1) and Delta5 desaturase in streptozotocin diabetic rats. Glargine insulin depressed the hyperglycemia of diabetic rats at 4h, but not after 24h of injection. Fenofibrate increased the radioimmunoreactive insulinemia in non-diabetic rats without changing the glycemia. Insulin increased the mRNAs and activities of SCD-1 and Delta5 desaturase depressed in diabetic rats. Fenofibrate increased acyl-CoA oxidase activity, and the mRNAs and activities of both desaturating enzymes in non-diabetic, diabetic and insulin-treated diabetic rats, but was less effective in the mRNAs modification of diabetic animals. Therefore, insulin, and fenofibrate through PPAR-alpha activation, enhance liver mRNAs and activities of SCD-1 and Delta5 desaturases independently and synergistically through different mechanisms. Insulin and fenofibrate independently increased the 18:1/18:0 ratio in liver lipids, increasing the fluidity of the membranes. The 20:4/18:2 ratio was maintained. Fenofibrate increased palmitic acid, but decreased stearic acid percentage in liver lipids.


Subject(s)
Diabetes Mellitus, Experimental/blood , Fatty Acids, Unsaturated/biosynthesis , Fenofibrate/administration & dosage , Insulin/administration & dosage , Insulin/blood , Acyl-CoA Oxidase/drug effects , Acyl-CoA Oxidase/metabolism , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Delta-5 Fatty Acid Desaturase , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/enzymology , Disease Models, Animal , Fatty Acid Desaturases/drug effects , Fatty Acid Desaturases/metabolism , Insulin/analogs & derivatives , Lipids/chemistry , Liver/chemistry , Liver/enzymology , Liver/metabolism , Male , RNA, Messenger/drug effects , Rats , Rats, Wistar , Stearoyl-CoA Desaturase/drug effects , Stearoyl-CoA Desaturase/metabolism , Streptozocin
7.
Nutrition ; 21(4): 512-9, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15811773

ABSTRACT

OBJECTIVE: The present work was designed to study the effects of the two main isomers of conjugated linoleic acid (CLA), cis-9,trans-11 and trans-10,cis-12, on liver composition and hepatic fatty acid oxidation in hamsters. METHODS: Animals were divided into three groups that were fed atherogenic diets supplemented with 0.5% linoleic acid, cis-9,trans-11 CLA, or trans-10,cis-12 CLA for 6 wk. Liver lipids, protein, water and DNA contents, and histologic structure were analyzed. Hepatic carnitine palmitoyltransferase-I and acyl coenzyme A oxidase activities were assessed. Triacylglycerol concentration, and aspartate aminotransferase, alanine aminotransferase, gamma-glutamyltransferase, and alkaline phosphatase activities were evaluated in serum. CLA isomer contents were analyzed by gas chromatography in hepatic triacylglycerols. Peroxisome proliferator-activated receptor-alpha mRNA was determined by reverse transcriptase polymerase chain reaction. RESULTS: Trans-10,cis-12 CLA led to significantly greater weight, lower levels of triacylglycerol, cholesterol, and phospholipid, and larger total cell number in liver. Carnitine palmitoyltransferase-I and acyl coenzyme A oxidase activities were significantly increased by this isomer. No changes were induced by cis-9,trans-11 CLA. Trans-10,cis-12 CLA was recovered in significantly lower proportions than cis-9,trans-11 in liver triacylglycerols. Histopathologic analysis showed no abnormalities. No significant differences in serum aspartate aminotransferase, alanine aminotransferase, gamma-glutamyltransferase, and alkaline phosphatase activities or in hepatic mRNA peroxisome proliferator-activated receptor-alpha expression were found among the three experimental groups. CONCLUSIONS: These results suggest that the addition of 0.5% of these CLA isomers to the diet do not induce toxic effects in liver after 6 wk of feeding. Intake of trans-10,cis-12 isomer but not of cis-9,trans-11 CLA increases liver fatty acid oxidation. This effect leads to decreased hepatic and serum triacylglycerols.


Subject(s)
Fatty Acids/metabolism , Linoleic Acids, Conjugated/pharmacology , Liver/drug effects , Liver/metabolism , Acyl-CoA Oxidase/drug effects , Acyl-CoA Oxidase/metabolism , Alkaline Phosphatase/blood , Animals , Body Weight/drug effects , Carnitine O-Palmitoyltransferase/drug effects , Carnitine O-Palmitoyltransferase/metabolism , Chromatography, Gas/methods , Cricetinae , DNA/drug effects , DNA/metabolism , Isomerism , Lipid Metabolism , Liver/enzymology , Male , Organ Size/drug effects , Oxidation-Reduction/drug effects , Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Time Factors , Transferases/blood , Triglycerides/blood , Water/metabolism
8.
Carcinogenesis ; 26(1): 219-27, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15447978

ABSTRACT

Prolonged administration of peroxisome proliferators to rodents typically leads to hepatocarcinogenesis. Peroxisome proliferator-activated receptor-alpha (PPARalpha) is required to mediate alterations in PPARalpha target gene expression, repress apoptosis, enhance replicative DNA synthesis, oxidative stress to DNA and hepatocarcinogenesis induced by the relatively specific PPARalpha agonist, Wy-14,643. Interestingly, administration of the less specific PPARalpha agonist, bezafibrate, leads to a modest induction of PPARalpha target genes in the absence of PPARalpha expression. In these studies, the role of PPARalpha in modulating hepatocarcinogenesis induced by long-term feeding of 0.5% bezafibrate was examined in wild-type (+/+) and PPARalpha-null (-/-) mice. The average liver weight was significantly higher in (+/+) and (-/-) mice fed bezafibrate than controls, but this effect was considerably less in (-/-) mice as compared with similarly treated (+/+) mice. Increased levels of mRNA encoding cell cycle regulatory proteins and DNA repair enzymes were found in (+/+) mice fed bezafibrate, and this effect was not found in (-/-) mice. In mice fed bezafibrate for 1 year, preneoplastic foci, adenomas and a hepatocellular carcinoma were found in (+/+) mice, while only a single microscopic adenoma was found in one (-/-) mouse. This effect was observed in both Sv/129 and C57BL/6N strains of mice, although only preneoplastic foci were observed in the latter strain. Interestingly, hepatic cholestasis was observed in 100% of the bezafibrate-fed (-/-) mice, and this was accompanied by significantly elevated hepatic expression of mRNA encoding bile salt export pump and lower expression of mRNA encoding cytochrome P450 7A1, consistent with enhanced activation of the bile acid receptor, farnesoid X receptor. Results from these studies demonstrate that the PPARalpha is required to mediate hepatocarcinogenesis induced by bezafibrate, and that PPARalpha protects against potential cholestasis.


Subject(s)
Bezafibrate/metabolism , Cholestasis/chemically induced , Liver Neoplasms/metabolism , PPAR alpha/deficiency , Peroxisome Proliferators/toxicity , Acyl-CoA Oxidase/drug effects , Acyl-CoA Oxidase/metabolism , Animals , Bezafibrate/toxicity , Bile Acids and Salts/metabolism , Blotting, Northern , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cholestasis/metabolism , Cholestasis/pathology , Cytochrome P-450 CYP4A/drug effects , Cytochrome P-450 CYP4A/metabolism , DNA Repair Enzymes/drug effects , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Disease Models, Animal , Liver Neoplasms/chemically induced , Liver Neoplasms/pathology , Male , Mice , PPAR alpha/genetics , RNA, Messenger/analysis , Receptors, Cytoplasmic and Nuclear , Transcription Factors/drug effects , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...