Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 21(4)2020 Feb 11.
Article in English | MEDLINE | ID: mdl-32054087

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a chronic disease in which excessive amount of lipids is accumulated as droplets in hepatocytes. Recently, cumulative evidences suggested that a sustained de novo lipogenesis can play an important role in NAFLD. Dysregulated expression of lipogenic genes, including ATP-citrate lyase (ACLY), has been found in liver diseases associated with lipid accumulation. ACLY is a ubiquitous cytosolic enzyme positioned at the intersection of nutrients catabolism and cholesterol and fatty acid biosyntheses. In the present study, the molecular mechanism of ACLY expression in a cell model of steatosis has been reported. We identified an internal ribosome entry site (IRES) in the 5' untranslated region of the ACLY mRNA, that can support an efficient mRNA translation through a Cap-independent mechanism. In steatotic HepG2 cells, ACLY expression was up-regulated through IRES-mediated translation. Since it has been demonstrated that lipid accumulation in cells induces endoplasmic reticulum (ER) stress, the involvement of this cellular pathway in the translational regulation of ACLY has been also evaluated. Our results showed that ACLY expression was increased in ER-stressed cells, through IRES-mediated translation of ACLY mRNA. A potential role of the Cap-independent translation of ACLY in NAFLD has been discussed.


Subject(s)
ATP Citrate (pro-S)-Lyase/genetics , Hepatocytes/metabolism , Lipogenesis , Protein Biosynthesis , RNA, Messenger/genetics , 5' Untranslated Regions , ATP Citrate (pro-S)-Lyase/metabolism , Endoplasmic Reticulum Stress , Hep G2 Cells , Humans , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , RNA, Messenger/metabolism
2.
Molecules ; 25(1)2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31906370

ABSTRACT

l-Carnitine is an amino acid derivative widely known for its involvement in the transport of long-chain fatty acids into the mitochondrial matrix, where fatty acid oxidation occurs. Moreover, l-Carnitine protects the cell from acyl-CoA accretion through the generation of acylcarnitines. Circulating carnitine is mainly supplied by animal-based food products and to a lesser extent by endogenous biosynthesis in the liver and kidney. Human muscle contains high amounts of carnitine but it depends on the uptake of this compound from the bloodstream, due to muscle inability to synthesize carnitine. Mitochondrial fatty acid oxidation represents an important energy source for muscle metabolism particularly during physical exercise. However, especially during high-intensity exercise, this process seems to be limited by the mitochondrial availability of free l-carnitine. Hence, fatty acid oxidation rapidly declines, increasing exercise intensity from moderate to high. Considering the important role of fatty acids in muscle bioenergetics, and the limiting effect of free carnitine in fatty acid oxidation during endurance exercise, l-carnitine supplementation has been hypothesized to improve exercise performance. So far, the question of the role of l-carnitine supplementation on muscle performance has not definitively been clarified. Differences in exercise intensity, training or conditioning of the subjects, amount of l-carnitine administered, route and timing of administration relative to the exercise led to different experimental results. In this review, we will describe the role of l-carnitine in muscle energetics and the main causes that led to conflicting data on the use of l-carnitine as a supplement.


Subject(s)
Carnitine/analogs & derivatives , Carnitine/metabolism , Energy Metabolism/drug effects , Fatty Acids/metabolism , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Carnitine/administration & dosage , Carnitine/biosynthesis , Carnitine/chemistry , Carnitine/pharmacology , Carnitine O-Palmitoyltransferase/metabolism , Dietary Supplements/adverse effects , Exercise/physiology , Humans , Methylamines/metabolism , Muscle, Skeletal/drug effects , Oxidation-Reduction
3.
Int J Biochem Cell Biol ; 117: 105618, 2019 12.
Article in English | MEDLINE | ID: mdl-31542428

ABSTRACT

Quercetin (Que), a widely distributed flavonoid in the human diet, exerts neuroprotective action because of its property to antagonize oxidative stress. Here, we investigated the effects of Que on lipid synthesis in C6 glioma cells. A rapid Que-induced inhibition of cholesterol and, to a lesser extent, of fatty acid synthesis from [1-14C]acetate was observed. The maximum decrease was detected at the level of palmitate, the end product of de novo fatty acid synthesis. The effect of Que on the enzyme activities of acetyl-CoA carboxylase 1 (ACC1) and fatty acid synthase (FAS), the two enzymes of this pathway, was investigated directly in situ in permeabilized C6 cells. An inhibitory effect on ACC1 was observed after 4 h of 25 µM Que treatment, while FAS activity was not affected. A reduction of polar lipid biosynthesis was also detected. A remarkable decrease of 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) activity, regulatory enzyme of cholesterol synthesis, was evidenced. Expression studies demonstrated that Que acts at transcriptional level, by reducing the mRNA abundance and protein amount of ACC1 and HMGCR. Deepening the molecular mechanism, we found that Que decreased the expression of SREBP-1 and SREBP-2, transcriptional factors representing the main regulators of de novo fatty acid and cholesterol synthesis, respectively. Que also reduced the nuclear content of ChREBP, a glucose-induced transcription factor involved in the regulation of lipogenic genes. Our results represent the first evidence that a direct and rapid downregulatory effect of Que on cholesterol and de novo fatty acid synthesis is elicited in C6 cells.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cholesterol/metabolism , Fatty Acids/metabolism , Glioma/metabolism , Quercetin/antagonists & inhibitors , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Humans , Rats
4.
FASEB J ; 33(1): 1428-1439, 2019 01.
Article in English | MEDLINE | ID: mdl-30133327

ABSTRACT

It is widely accepted that chronic stress may alter the homeostatic mechanisms of body weight control. In this study, we followed the metabolic changes occurring in mice when chronic stress caused by psychosocial defeat (CPD) is associated with ad libitum exposure to a palatable high-fat diet (HFD). In this model, CPD mice consumed more HFD than unstressed (Un) mice without gaining body weight. We focused on metabolic processes involved in weight control, such as de novo lipogenesis (DNL), fatty acid ß-oxidation (FAO), and thermogenesis. The activity and expression of DNL enzymes were reduced in the liver and white adipose tissue of mice consuming the HFD. Such effects were particularly evident in stressed mice. In both CPD and Un mice, HFD consumption increased the hepatic expression of the mitochondrial FAO enzyme carnitine palmitoyltransferase-1. In the liver of mice consuming the HFD, stress exposure prevented accumulation of triacylglycerols; however, accumulation of triacylglycerols was observed in Un mice under the same dietary regimen. In brown adipose tissue, stress increased the expression of uncoupling protein-1, which is involved in energy dissipation, both in HFD and control diet-fed mice. We consider increased FAO and energy dissipation responsible for the antiobesity effect seen in CPD/HFD mice. However, CPD associated with HFD induced hepatic oxidative stress.-Giudetti, A. M., Testini, M., Vergara, D., Priore, P., Damiano, F., Gallelli, C. A., Romano, A., Villani, R., Cassano, T., Siculella, L., Gnoni, G. V., Moles, A., Coccurello, R., Gaetani, S. Chronic psychosocial defeat differently affects lipid metabolism in liver and white adipose tissue and induces hepatic oxidative stress in mice fed a high-fat diet.


Subject(s)
Adipose Tissue, White/metabolism , Diet, High-Fat , Lipid Metabolism , Liver/metabolism , Oxidative Stress , Stress, Psychological , Acetyl-CoA Carboxylase/metabolism , Adipose Tissue, Brown/enzymology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/enzymology , Animals , Body Weight , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Disease Models, Animal , Energy Intake , Fatty Acid Synthases/metabolism , Fatty Acids/metabolism , Glutathione/metabolism , Liver/enzymology , Male , Mice , Mice, Inbred BALB C , RNA, Messenger/genetics , Uncoupling Protein 1/metabolism
5.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(4): 388-398, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29343429

ABSTRACT

Acetyl-CoA carboxylase 1 (ACC1) is a cytosolic enzyme catalyzing the rate limiting step in de novo fatty acid biosynthesis. There is mounting evidence showing that ACC1 is susceptible to dysregulation and that it is over-expressed in liver diseases associated with lipid accumulation and in several cancers. In the present study, ACC1 regulation at the translational level is reported. Using several experimental approaches, the presence of an internal ribosome entry site (IRES) has been established in the 5' untranslated region (5' UTR) of the ACC1 mRNA. Transfection experiments with the ACC1 5' UTR inserted in a dicistronic reporter vector show a remarkable increase in the downstream cistron translation, through a cap-independent mechanism. The endoplasmic reticulum (ER) stress condition and the related unfolded protein response (UPR), triggered by treatment with thapsigargin and tunicamycin, cause an increase of the cap-independent translation of ACC1 mRNA in HepG2 cells, despite the overall reduction in global protein synthesis. Other stress conditions, such as serum starvation and incubation with hypoxia mimetic agent CoCl2, up-regulate ACC1 expression in HepG2 cells at the translational level. Overall, these findings indicate that the presence of an IRES in the ACC1 5' UTR allows ACC1 mRNA translation in conditions that are inhibitory to cap-dependent translation. A potential involvement of the cap-independent translation of ACC1 in several pathologies, such as obesity and cancer, has been discussed.


Subject(s)
Acetyl-CoA Carboxylase/genetics , Cobalt/pharmacology , Endoplasmic Reticulum Stress/drug effects , Internal Ribosome Entry Sites/genetics , Protein Biosynthesis , 5' Untranslated Regions/genetics , Acetyl-CoA Carboxylase/metabolism , Cell Hypoxia/drug effects , Cell Survival/drug effects , Culture Media, Serum-Free , Hep G2 Cells , Humans , Plasmids/metabolism , Protein Biosynthesis/drug effects , RNA Splicing/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Oxid Med Cell Longev ; 2017: 9076052, 2017.
Article in English | MEDLINE | ID: mdl-29435099

ABSTRACT

Recently, the discovery of natural compounds capable of modulating nervous system function has revealed new perspectives for a healthier brain. Here, we investigated the effects of oleic acid (OA) and hydroxytyrosol (HTyr), two important extra virgin olive oil compounds, on lipid synthesis in C6 glioma cells. OA and HTyr inhibited both de novo fatty acid and cholesterol syntheses without affecting cell viability. The inhibitory effect of the individual compounds was more pronounced if OA and HTyr were administered in combination. A reduction of polar lipid biosynthesis was also detected, while triglyceride synthesis was marginally affected. To clarify the lipid-lowering mechanism of these compounds, their effects on the activity of key enzymes of fatty acid biosynthesis (acetyl-CoA carboxylase-ACC and fatty acid synthase-FAS) and cholesterologenesis (3-hydroxy-3-methylglutaryl-CoA reductase-HMGCR) were investigated in situ by using digitonin-permeabilized C6 cells. ACC and HMGCR activities were especially reduced after 4 h of 25 µM OA and HTyr treatment. No change in FAS activity was observed. Inhibition of ACC and HMGCR activities is corroborated by the decrease of their mRNA abundance and protein level. Our results indicate a direct and rapid downregulatory effect of the two olive oil compounds on lipid synthesis in C6 cells.


Subject(s)
Anticholesteremic Agents/pharmacology , Cholesterol/metabolism , Fatty Acid Synthases/antagonists & inhibitors , Glioma/metabolism , Oleic Acid/pharmacology , Phenylethyl Alcohol/analogs & derivatives , Animals , Cell Line, Tumor , Fatty Acid Synthases/metabolism , Lipid Metabolism/drug effects , Olive Oil/chemistry , Phenylethyl Alcohol/pharmacology , Rats
7.
Int J Mol Sci ; 17(6)2016 May 25.
Article in English | MEDLINE | ID: mdl-27231907

ABSTRACT

The transport of solutes across the inner mitochondrial membrane is catalyzed by a family of nuclear-encoded membrane-embedded proteins called mitochondrial carriers (MCs). The citrate carrier (CiC) and the carnitine/acylcarnitine transporter (CACT) are two members of the MCs family involved in fatty acid metabolism. By conveying acetyl-coenzyme A, in the form of citrate, from the mitochondria to the cytosol, CiC contributes to fatty acid and cholesterol synthesis; CACT allows fatty acid oxidation, transporting cytosolic fatty acids, in the form of acylcarnitines, into the mitochondrial matrix. Fatty acid synthesis and oxidation are inversely regulated so that when fatty acid synthesis is activated, the catabolism of fatty acids is turned-off. Malonyl-CoA, produced by acetyl-coenzyme A carboxylase, a key enzyme of cytosolic fatty acid synthesis, represents a regulator of both metabolic pathways. CiC and CACT activity and expression are regulated by different nutritional and hormonal conditions. Defects in the corresponding genes have been directly linked to various human diseases. This review will assess the current understanding of CiC and CACT regulation; underlining their roles in physio-pathological conditions. Emphasis will be placed on the molecular basis of the regulation of CiC and CACT associated with fatty acid metabolism.


Subject(s)
Carnitine Acyltransferases/metabolism , Carrier Proteins/metabolism , Cholesterol/biosynthesis , Fatty Acids/biosynthesis , Thyroid Hormones/physiology , Acetyl Coenzyme A/metabolism , Animals , Carnitine Acyltransferases/genetics , Carrier Proteins/genetics , Cytosol/metabolism , Gene Expression Regulation, Enzymologic , Humans , Lipogenesis , Mitochondria/metabolism
8.
IUBMB Life ; 67(1): 9-17, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25631376

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) represents the most common chronic liver disease in western countries, being considered the hepatic manifestation of metabolic syndrome. Cumulative lines of evidence suggest that olive oil, used as primary source of fat by Mediterranean populations, may play a key role in the observed health benefits on NAFLD. In this review, we summarize the state of the art of the knowledge on the protective role of both major and minor components of olive oil on lipid metabolism during NAFLD. In particular, the biochemical mechanisms responsible for the increase or decrease in hepatic lipid content are critically analyzed, taking into account that several studies have often provided different and/or conflicting results in animal models fed on olive oil-enriched diet. In addition, new findings that highlight the hypolipidemic and the antisteatotic actions of olive oil phenols are presented. As mitochondrial dysfunction plays a key role in the pathogenesis of NAFLD, the targeting of these organelles with olive oil phenols as a powerful therapeutic approach is also discussed.


Subject(s)
Lipid Metabolism/drug effects , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Olive Oil/chemistry , Phenols/pharmacology , Humans , Lipid Metabolism/physiology , Mitochondria/drug effects , Olive Oil/pharmacology
9.
Eur J Nutr ; 54(5): 823-33, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25296884

ABSTRACT

PURPOSE: Regular consumption of extra virgin olive oil (EVOO) is associated with a low incidence of atherosclerotic diseases. The phenolic component contributes to the hypolipidemic action of EVOO, although the biochemical mechanisms leading this beneficial outcome are not fully understood. Since liver plays a pivotal role in the whole body lipid homeostasis, we investigated the short-term effects of EVOO extract, with a high phenol content (HPE), on lipid synthesis in primary rat hepatocytes. Refined olive oil extract, with a low phenol content, was used throughout this study as a control. METHODS: Olive oil phenols isolated with methanolic extractions were subsequently analyzed by high performance liquid chromatography, electrospray ionization tandem mass spectrometry, and gas chromatography mass spectrometry. Rat hepatocytes were obtained from collagenase perfusion of liver. A colorimetric assay was performed to exclude cytotoxicity of the extracts. Radioenzymatic methods were used in order to investigate hepatic lipid metabolism. RESULTS: HPE, dose- (0.1-50 µg/mL) and time-dependently (0.5-4 h) inhibited both lipogenesis and cholesterogenesis (n = 6, P < 0.05), as well as triglycerides synthesis (n = 5, P < 0.05). We showed that these effects are attributable to a short-term modulation by HPE of the key enzymes implicated in the abovementioned pathways (n = 5, P < 0.05). CONCLUSIONS: The decrease in hepatic lipid synthesis may represent a potential mechanism underlying the hypolipidemic effect of EVOO phenols.


Subject(s)
Lipid Metabolism/drug effects , Liver/drug effects , Olive Oil/chemistry , Phenols/pharmacology , Animals , Cell Survival/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid , Down-Regulation , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/cytology , Liver/metabolism , Male , Rats , Rats, Wistar , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
10.
Biochim Biophys Acta ; 1831(12): 1679-89, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23948263

ABSTRACT

Thyroid hormone 3,5,3'-triiodo-l-thyronine (T3) is known to affect cell metabolism through both the genomic and non-genomic actions. Recently, we demonstrated in HepG2 cells that T3 controls the expression of SREBP-1, a transcription factor involved in the regulation of lipogenic genes. This occurs by activation of a cap-independent translation mechanism of its mRNA. Such a process is dependent on non-genomic activation of both MAPK/ERK and PI3K/Akt pathways. The physiological role of 3,5-diiodo-l-thyronine (T2), previously considered only as a T3 catabolite, is of growing interest. Evidences have been reported that T2 rapidly affects some metabolic pathways through non-genomic mechanisms. Here, we show that T2, unlike T3, determines the block of proteolytic cleavage of SREBP-1 in HepG2 cells, without affecting its expression at the transcriptional or translational level. Consequently, Fatty Acid Synthase expression is reduced. T2 effects depend on the concurrent activation of MAPKs ERK and p38, of Akt and PKC-δ pathways. Upon the activation of these signals, apoptosis of HepG2 cells seems to occur, starting at 12h of T2 treatment. PKC-δ appears to act as a switch between p38 activation and Akt suppression, suggesting that this PKC may function as a controller in the balance of pro-apoptotic (p38) and anti-apoptotic (Akt) signals in HepG2 cells.


Subject(s)
Apoptosis/drug effects , Diiodothyronines/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , Proteolysis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Sterol Regulatory Element Binding Protein 1/genetics , Triiodothyronine/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Int J Biochem Cell Biol ; 44(4): 659-68, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22249025

ABSTRACT

Citrate carrier (CiC), a mitochondrial inner membrane protein, is an essential component of the shuttle system which transports acetyl-CoA from mitochondria to the cytosol where lipogenesis occurs. CiC is regulated by SREBP-1, a transcription factor that controls the expression of several lipogenic genes. CiC is also implicated in cholesterol synthesis, glycolysis and gluconeogenesis, suggesting that besides SREBP-1 other transcription factors could modulate the expression of its gene. Here, we provide evidences demonstrating that CiC expression is regulated by peroxisome proliferator-activated receptor (PPAR) alpha and gamma in hepatocytes and adipocytes, respectively. CiC expression increased in rat BRL-3A hepatocytes treated with WY-14,643, agonist of PPARα, and in murine 3T3-L1 adipocytes treated with rosiglitazone, agonist of PPARγ. The overexpression of PPARα/RXRα and PPARγ/RXRα heterodimer enhanced CiC promoter activity in BRL-3A and 3T3-L1, respectively. Luciferase reporter gene and gel mobility shift assays indicated that a functional peroxisome proliferator-activated receptor response element (PPRE), identified in the CiC promoter, conferred responsiveness to activation by PPARs. The binding of PPRE of CiC promoter by PPARα and PPARγin vivo was confirmed by ChIP assay in BRL-3A and 3T3-L1 cells, respectively.


Subject(s)
Adipocytes/metabolism , Carrier Proteins/genetics , Hepatocytes/metabolism , PPAR alpha/metabolism , PPAR gamma/metabolism , Promoter Regions, Genetic/genetics , Adipocytes/cytology , Adipocytes/drug effects , Animals , Base Sequence , Cell Differentiation/drug effects , Docosahexaenoic Acids/pharmacology , Genes, Reporter/genetics , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Luciferases/genetics , Mice , Promoter Regions, Genetic/drug effects , Protein Binding , Rats , Response Elements/drug effects , Response Elements/genetics , Transcriptional Activation/drug effects
12.
J Cell Physiol ; 227(6): 2388-97, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21826653

ABSTRACT

Liver is an important target for thyroid hormone actions. T(3) exerts its effects by two mechanisms: (i) Genomic actions consisting of T(3) link to nuclear receptors that bind responsive elements in the promoter of target genes, (ii) non-genomic actions including integrin αvb3 receptor-mediated MAPK/ERK and PI3K/Akt/mTOR-C1 activation. SREBP-1a, SREBP-1c, and SREBP-2 are transcription factors involved in the regulation of lipogenic genes. We show in Hep G2 cells that T(3) determined a dose- and time-dependent increase in the level of the precursor form of SREBP-1 without affecting SREBP-1 mRNA abundance. T(3) also induced phosphorylation of ERK1/2, Akt and of mTOR-C1 target S6K-P70, and the cytosol-to-membrane translocation of PKC-α. Modulation of SREBP-1 protein level by T(3) was dependent on MAPK/ERK, PI3K/Akt/mTOR-C1 pathway activation since the MEK inhibitor PD98059 or the PI3K inhibitor LY294002 abolished the stimulatory effect of T(3) . Conversely, the effect of T(3) on SREBP-1 level was enhanced by using rapamycin, mTOR-C1 inhibitor. These data suggest a negative control of mTOR-C1 target S6K-P70 on PI3K/Akt pathway. The effect of T(3) on SREBP-1 content increased also by using PKC inhibitors. These inhibitors increased the action of T(3) on Akt phosphorylation suggesting that conventional PKCs may work as negative regulators of the T(3) -dependent SREBP-1 increase. T(3) effects were partially abrogated by tetrac, an inhibitor of the T(3) -αvß3 receptor interaction and partially evoked by T(3) analog T(3) -agarose. These findings support a model in which T(3) activates intracellular signaling pathways which may be involved in the increment of SREBP-1 level through an IRES-mediated translation mechanism.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Signal Transduction , Sterol Regulatory Element Binding Protein 1/metabolism , Triiodothyronine/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Feedback, Physiological , Hep G2 Cells , Humans , Integrin alphaVbeta3/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mechanistic Target of Rapamycin Complex 1 , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Multiprotein Complexes , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Protein Kinase C-alpha/metabolism , Protein Kinase Inhibitors/pharmacology , Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , Sterol Regulatory Element Binding Protein 1/genetics , TOR Serine-Threonine Kinases , Time Factors , Up-Regulation
13.
PLoS One ; 6(9): e24084, 2011.
Article in English | MEDLINE | ID: mdl-21909411

ABSTRACT

There is growing evidence that mitochondrial dysfunction, and more specifically fatty acid ß-oxidation impairment, is involved in the pathophysiology of non-alcoholic steatohepatitis (NASH). The goal of the present study was to achieve more understanding on the modification/s of carnitinepalmitoyltransferase-I (CPT-I), the rate-limiting enzyme of the mitochondrial fatty acid ß-oxidation, during steatohepatitis. A high fat/methionine-choline deficient (MCD) diet, administered for 4 weeks, was used to induce NASH in rats.We demonstrated that CPT-I activity decreased, to the same extent, both in isolated liver mitochondria and in digitonin-permeabilized hepatocytes from MCD-diet fed rats.At the same time, the rate of total fatty acid oxidation to CO(2) and ketone bodies, measured in isolated hepatocytes, was significantly lowered in treated animals when compared to controls. Finally, an increase in CPT-I mRNA abundance and protein content, together with a high level of CPT-I protein oxidation was observed in treated rats. A posttranslational modification of rat CPT-I during steatohepatitis has been here discussed.


Subject(s)
Carnitine O-Palmitoyltransferase/metabolism , Choline/pharmacology , Diet , Fatty Acids/metabolism , Feeding Behavior/drug effects , Liver/enzymology , Methionine/pharmacology , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Acetyl-CoA Carboxylase/metabolism , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Carnitine O-Palmitoyltransferase/genetics , Cell Membrane Permeability/drug effects , Choline/administration & dosage , Enzyme Assays , Fatty Acids/blood , Gene Expression Regulation, Enzymologic/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/drug effects , Liver/pathology , Male , Malonyl Coenzyme A/pharmacology , Methionine/administration & dosage , Methionine/deficiency , Mitochondria, Liver/drug effects , Mitochondria, Liver/enzymology , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Oxidation-Reduction/drug effects , Palmitic Acid/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar
14.
Biochem J ; 429(3): 603-12, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20513236

ABSTRACT

SREBPs (sterol-regulatory-element-binding proteins) are a family of transcription factors that modulate the expression of several enzymes implicated in endogenous cholesterol, fatty acid, triacylglycerol and phospholipid synthesis. In the present study, evidence for SREBP-1 regulation at the translational level is reported. Using several experimental approaches, we have demonstrated that the 5'-UTR (untranslated region) of the SREBP-1a mRNA contains an IRES (internal ribosome entry site). Transfection experiments with the SREBP-1a 5'-UTR inserted in a dicistronic reporter vector showed a remarkable increase in the downstream cistron translation, through a cap-independent mechanism. Insertion of the SREBP-1c 5'-UTR in the same vector also stimulated the translation of the downstream cistron, but the observed effect can be ascribed, at least in part, to a cryptic promoter activity. Cellular stress conditions, such as serum starvation, caused an increase in the level of SREBP-1 precursor and mature form in both Hep G2 and HeLa cells, despite the overall reduction in protein synthesis, whereas mRNA levels for SREBP-1 were unaffected by serum starvation. Transfection experiments carried out with a dicistronic construct demonstrated that the cap-dependent translation was affected more than IRES-mediated translation by serum starvation. The thapsigargin- and tunicamycin-induced UPR (unfolded protein response) also increased SREBP-1 expression in Hep G2 cells, through the cap-independent translation mediated by IRES. Overall, these findings indicate that the presence of IRES in the SREBP-1a 5'-UTR allows translation to be maintained under conditions that are inhibitory to cap-dependent translation.


Subject(s)
Protein Biosynthesis , RNA, Messenger/genetics , Ribosomes/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , 5' Untranslated Regions , Base Sequence , Cell Line , Culture Media, Serum-Free , DNA Primers , Genes, Reporter , Humans
15.
Mol Microbiol ; 77(3): 716-29, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20545843

ABSTRACT

With the beginning of the idiophase the highly phosphorylated guanylic nucleotides guanosine 5'-diphosphate 3'-diphosphate (ppGpp) and guanosine 5'-triphosphate 3'-diphosphate (pppGpp), collectively referred to as (p)ppGpp, activate stress survival adaptation programmes and trigger secondary metabolism in actinomycetes. The major target of (p)ppGpp is the RNA polymerase, where it binds altering the enzyme activity. In this study analysis of the polynucleotide phosphorylase (PNPase)-encoding gene pnp mRNA, in Nonomuraea sp. ATCC 39727 wild-type, constitutively stringent and relaxed strains, led us to hypothesize that in actinomycetes (p)ppGpp may modulate gene expression at the level of RNA decay also. This hypothesis was supported by: (i) in vitro evidence that ppGpp, at physiological levels, inhibited both polynucleotide polymerase and phosphorolytic activities of PNPase in Nonomuraea sp., but not in Escherichia coli, (ii) in vivo data showing that the pnp mRNA and the A40926 antibiotic cluster-specific dpgA mRNA were stabilized during the idiophase in the wild-type strain but not in a relaxed mutant and (iii) measurement of chemical decay of pulse-labelled bulk mRNA. The results of biochemical tests suggest competitive inhibition of ppGpp with respect to nucleoside diphosphates in polynucleotide polymerase assays and mixed inhibition with respect to inorganic phosphate when the RNA phosphorolytic activity was determined.


Subject(s)
Actinobacteria/enzymology , Bacterial Proteins/metabolism , Down-Regulation , Gene Expression Regulation, Enzymologic , Guanosine Tetraphosphate/metabolism , Polyribonucleotide Nucleotidyltransferase/metabolism , Actinobacteria/genetics , Actinobacteria/metabolism , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Polyribonucleotide Nucleotidyltransferase/genetics
16.
Endocrinology ; 151(4): 1551-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20203153

ABSTRACT

Citrate carrier (CiC), an integral protein of the mitochondrial inner membrane, plays an important role in hepatic intermediary metabolism, supplying the cytosol with acetyl-coenzyme A for fatty acid and cholesterol synthesis. Here, the effect of streptozotocin-induced diabetes on CiC activity and expression in rat liver was investigated. The rate of citrate transport was reduced by about 35% in mitochondria from diabetic vs. control rats. Kinetic studies in mitochondria from diabetic rats showed a reduction in maximum velocity and almost unchanged Michaelis-Menten constant of the CiC protein. Mitochondrial phospholipid amount was not significantly affected, whereas an increase in the cholesterol content and in the cholesterol/phospholipid ratio was observed. To thoroughly investigate the mechanism responsible for the reduced CiC activity in the diabetic state, molecular studies were performed. Ribonuclease protection assays and Western blotting analysis indicated that both hepatic CiC mRNA accumulation and protein level decreased similarly to the CiC activity. The reduced mRNA level and the lower content of the mitochondrial CiC protein, might account for the decline of CiC activity in diabetic animals. To discriminate between the role played by hyperglycemia from that of hypoinsulinemia in the reduction of CiC activity and expression, studies were conducted administrating phlorizin or insulin to streptozotocin-diabetic rats. Our data indicated that both insulin and glucose affect CiC activity and expression in diabetic rats, although they act at different regulatory steps.


Subject(s)
Carrier Proteins/metabolism , Citric Acid/metabolism , Diabetes Mellitus, Experimental/metabolism , Liver/metabolism , Mitochondria, Liver/metabolism , Analysis of Variance , Animals , Blood Glucose/metabolism , Blotting, Western , Body Weight/drug effects , Body Weight/physiology , Carrier Proteins/genetics , Cholesterol/blood , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Insulin/metabolism , Insulin/pharmacology , Liver/drug effects , Liver/pathology , Male , Mitochondria, Liver/drug effects , Mitochondria, Liver/genetics , Mitochondrial Membranes/metabolism , Organ Size/drug effects , Organ Size/physiology , Phlorhizin/metabolism , Phlorhizin/pharmacology , Phospholipids/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar
17.
Biochim Biophys Acta ; 1797(2): 233-40, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19878644

ABSTRACT

Besides triiodothyronine (T3), 3,5-diiodo-L-thyronine (T2) has been reported to affect mitochondrial bioenergetic parameters. T2 effects have been considered as independent of protein synthesis. Here, we investigated the effect of in vivo chronic T2 administration to hypothyroid rats on liver mitochondrial F(o)F(1)-ATP synthase activity and expression. T2 increased state 4 and state 3 oxygen consumption and raised ATP synthesis and hydrolysis, which were reduced in hypothyroid rats. Immunoblotting analysis showed that T2 up-regulated the expression of several subunits (alpha, beta, F(o)I-PVP and OSCP) of the ATP synthase. The observed increase of beta-subunit mRNA accumulation suggested a T2-mediated nuclear effect. Then, the molecular basis underlying T2 effects was investigated. Our results support the notion that the beta-subunit of ATP synthase is indirectly regulated by T2 through, at least in part, the activation of the transcription factor GA-binding protein/nuclear respiratory factor-2. These findings provide new insights into the T2 role on bioenergetic mechanisms.


Subject(s)
Diiodothyronines/pharmacology , GA-Binding Protein Transcription Factor/metabolism , Hypothyroidism/metabolism , Mitochondria, Liver/drug effects , Mitochondria, Liver/enzymology , Mitochondrial Proton-Translocating ATPases/metabolism , Animals , Chromatin Immunoprecipitation , GA-Binding Protein Transcription Factor/genetics , Hypothyroidism/drug therapy , Immunoblotting , Male , Membrane Potential, Mitochondrial , Oxygen Consumption , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
18.
IUBMB Life ; 61(10): 987-94, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19787704

ABSTRACT

The citrate carrier (CiC), a nuclear-encoded protein located in the mitochondrial inner membrane, is a member of the mitochondrial carrier family. CiC plays an important role in hepatic lipogenesis, which is responsible for the efflux of acetyl-CoA from the mitochondria to the cytosol in the form of citrate, the primer for fatty acid and cholesterol synthesis. In addition, CiC is a key component of the isocitrate-oxoglutarate and the citrate-malate shuttles. CiC has been purified from various species and its reconstituted function characterized as well as its cDNA isolated and sequenced. CiC mRNA and/or CiC protein levels are high in liver, pancreas, and kidney, but are low or absent in brain, heart, skeletal muscle, placenta, and lungs. A reduction of CiC activity was found in diabetic, hypothyroid, starved rats, and in rats fed on a polyunsaturated fatty acid (PUFA)-enriched diet. Molecular analysis suggested that the regulation of CiC activity occurs mainly through transcriptional and post-transcriptional mechanisms. This review begins with an assessment of the current understanding of CiC structural and biochemical characteristics, underlying the structure-function relationship. Emphasis will be placed on the molecular basis of the regulation of CiC activity in coordination with fatty acid synthesis.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation , Mitochondria/metabolism , Acetyl Coenzyme A/metabolism , Animals , Binding Sites/genetics , Carrier Proteins/genetics , Carrier Proteins/physiology , Citric Acid/metabolism , Cytosol/metabolism , Fatty Acids/biosynthesis , Fatty Acids, Unsaturated/metabolism , Forecasting , Kinetics , Lipogenesis , Liver/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mitochondria/genetics , Mitochondria, Liver/metabolism , Promoter Regions, Genetic , Protein Binding/genetics , Rats
19.
Biochem J ; 417(2): 561-71, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-18795892

ABSTRACT

CiC (citrate carrier), a mitochondrial membrane protein, plays an important metabolic role by transporting acetyl-CoA into the cytosol for fatty acid and cholesterol synthesis. Several studies showed that CiC activity and expression is regulated by dietary fatty acids. In the present study we report data on the structural and functional characterization of the 5'-flanking region of the rat Cic gene. By transient transfection assays in H4IIE rat hepatoma cells, a PUFA (polyunsaturated fatty acids) response region has been identified within the CiC promoter. A cluster of putative binding sites for several transcription factors, composed of a NF-Y (nuclear factor-Y) site, an E-box-like site, a SRE1 (sterol regulatory element 1)-like site and four Sp1 (stimulatory protein 1) sites, was localized in the promoter region. Luciferase reporter gene and gel mobility shift assays indicated that a functional E-box-like, essential to the basal CiC promoter activity, confers responsiveness to activation by SREBP (SRE-binding protein)-1c. This study provides evidence for SREBP-1c as a principal target for PUFA regulation of CiC transcription. In H4IIE cells, overexpression of nSREBP (nuclear SREBP)-1c over-rides arachidonic acid (C(20:4, n-6)) suppression, but does not prevent the repression by docosahexaenoic acid (C(22:6, n-3)). ChIP (chromatin immunoprecipitation) assays in H4IIE cells showed that docosahexaenoic acid affects the binding of NF-Y, Sp1 and SREBP-1 to the PUFA response region of CiC promoter, whereas arachidonic acid alters only the binding of SREBP-1. Our data show that PUFA inhibition of hepatic Cic gene transcription is mediated not only by the nuclear level of SREBP-1c, but also might involve a reduction in Sp1 and NF-Y DNA binding, suggesting differential mechanisms in the Cic gene regulation by different PUFA.


Subject(s)
Carrier Proteins/metabolism , Liver/metabolism , Promoter Regions, Genetic/genetics , Animals , Carrier Proteins/genetics , Cell Line , Fatty Acids, Unsaturated , Gene Expression Regulation , Male , Molecular Sequence Data , Protein Binding , RNA, Messenger/genetics , Rats , Rats, Wistar , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Transcription, Genetic/genetics
20.
Biochim Biophys Acta ; 1777(10): 1326-35, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18775409

ABSTRACT

Extracellular ATP formation from ADP and inorganic phosphate, attributed to the activity of a cell surface ATP synthase, has so far only been reported in cultures of some proliferating and tumoral cell lines. We now provide evidence showing the presence of a functionally active ecto-F(o)F(1)-ATP synthase on the plasma membrane of normal tissue cells, i.e. isolated rat hepatocytes. Both confocal microscopy and flow cytometry analysis show the presence of subunits of F(1) (alpha/beta and gamma) and F(o) (F(o)I-PVP(b) and OSCP) moieties of ATP synthase at the surface of rat hepatocytes. This finding is confirmed by immunoblotting analysis of the hepatocyte plasma membrane fraction. The presence of the inhibitor protein IF(1) is also detected on the hepatocyte surface. Activity assays show that the ectopic-ATP synthase can work both in the direction of ATP synthesis and hydrolysis. A proton translocation assay shows that both these mechanisms are accompanied by a transient flux of H(+) and are inhibited by F(1) and F(o)-targeting inhibitors. We hypothesise that ecto-F(o)F(1)-ATP synthase may control the extracellular ADP/ATP ratio, thus contributing to intracellular pH homeostasis.


Subject(s)
Extracellular Space/metabolism , Hepatocytes/enzymology , Mitochondrial Proton-Translocating ATPases , Adenosine Triphosphate/metabolism , Animals , Cell Membrane/chemistry , Cell Membrane/enzymology , Cells, Cultured , Hepatocytes/cytology , Male , Membrane Potential, Mitochondrial/physiology , Mitochondrial Proton-Translocating ATPases/chemistry , Mitochondrial Proton-Translocating ATPases/metabolism , Protein Subunits/chemistry , Protein Subunits/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...