Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters











Publication year range
1.
Nutrients ; 13(6)2021 Jun 17.
Article in English | MEDLINE | ID: mdl-34204572

ABSTRACT

Diet is a major driver of gut microbiota variation and plays a role in metabolic disorders, including metabolic syndrome (MS). Mycorrhized foods from symbiotic agriculture (SA) exhibit improved nutritional properties, but potential benefits have never been investigated in humans. We conducted a pilot interventional study on 60 adults with ≥ 1 risk factors for MS, of whom 33 consumed SA-derived fresh foods and 27 received probiotics over 30 days, with a 15-day follow-up. Stool, urine and blood were collected over time to explore changes in gut microbiota, metabolome, and biochemical, inflammatory and immunologic parameters; previous dietary habits were investigated through a validated food-frequency questionnaire. The baseline microbiota showed alterations typical of metabolic disorders, mainly an increase in Coriobacteriaceae and a decrease in health-associated taxa, which were partly reversed after the SA-based diet. Improvements were observed in metabolome, MS presence (two out of six subjects no longer had MS) or components. Changes were more pronounced with less healthy baseline diets. Probiotics had a marginal, not entirely favorable, effect, although one out of three subjects no longer suffered from MS. These findings suggest that improved dietary patterns can modulate the host microbiota and metabolome, counteracting the risk of developing MS.


Subject(s)
Agriculture , Diet , Gastrointestinal Microbiome , Metabolic Syndrome/microbiology , Adolescent , Adult , Aged , Diet, Healthy , Feces/microbiology , Female , Humans , Italy , Male , Metabolome , Middle Aged , Pilot Projects , Probiotics , Young Adult
2.
Article in English | MEDLINE | ID: mdl-33808259

ABSTRACT

In recent years, lipid metabolism has gained greater attention in several diseases including cancer. Dysregulation of fatty acid metabolism is a key component in breast cancer malignant transformation. In particular, de novo lipogenesis provides the substrate required by the proliferating tumor cells to maintain their membrane composition and energetic functions during enhanced growth. However, it appears that not all breast cancer subtypes depend on de novo lipogenesis for fatty acid replenishment. Indeed, while breast cancer luminal subtypes rely on de novo lipogenesis, the basal-like receptor-negative subtype overexpresses genes involved in the utilization of exogenous-derived fatty acids, in the synthesis of triacylglycerols and lipid droplets, and fatty acid oxidation. These metabolic differences are specifically associated with genomic and proteomic changes that can perturb lipogenic enzymes and related pathways. This behavior is further supported by the observation that breast cancer patients can be stratified according to their molecular profiles. Moreover, the discovery that extracellular vesicles act as a vehicle of metabolic enzymes and oncometabolites may provide the opportunity to noninvasively define tumor metabolic signature. Here, we focus on de novo lipogenesis and the specific differences exhibited by breast cancer subtypes and examine the functional contribution of lipogenic enzymes and associated transcription factors in the regulation of tumorigenic processes.


Subject(s)
Breast Neoplasms , Lipogenesis , Fatty Acids , Humans , Lipid Metabolism , Proteomics
4.
Nutrients ; 13(2)2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33513874

ABSTRACT

Oleoylethanolamide (OEA) is a naturally occurring bioactive lipid belonging to the family of N-acylethanolamides. A variety of beneficial effects have been attributed to OEA, although the greater interest is due to its potential role in the treatment of obesity, fatty liver, and eating-related disorders. To better clarify the mechanism of the antiadipogenic effect of OEA in the liver, using a lipidomic study performed by 1H-NMR, LC-MS/MS and thin-layer chromatography analyses we evaluated the whole lipid composition of rat liver, following a two-week daily treatment of OEA (10 mg kg-1 i.p.). We found that OEA induced a significant reduction in hepatic triacylglycerol (TAG) content and significant changes in sphingolipid composition and ceramidase activity. We associated the antiadipogenic effect of OEA to decreased activity and expression of key enzymes involved in fatty acid and TAG syntheses, such as acetyl-CoA carboxylase, fatty acid synthase, diacylglycerol acyltransferase, and stearoyl-CoA desaturase 1. Moreover, we found that both SREBP-1 and PPARγ protein expression were significantly reduced in the liver of OEA-treated rats. Our findings add significant and important insights into the molecular mechanism of OEA on hepatic adipogenesis, and suggest a possible link between the OEA-induced changes in sphingolipid metabolism and suppression of hepatic TAG level.


Subject(s)
Endocannabinoids/therapeutic use , Fatty Acids/metabolism , Liver/metabolism , Oleic Acids/therapeutic use , PPAR gamma/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/metabolism , Animals , Cell Line, Tumor , Chromatography, Liquid , Diacylglycerol O-Acyltransferase/metabolism , Lipogenesis , Magnetic Resonance Spectroscopy , Male , Multivariate Analysis , Rats , Rats, Wistar , Stearoyl-CoA Desaturase/metabolism , Tandem Mass Spectrometry
5.
Front Neurosci ; 14: 783, 2020.
Article in English | MEDLINE | ID: mdl-32792906

ABSTRACT

Medium-chain fatty acids (MCFA) are dietary components with a chain length ranging from 6 to 12 carbon atoms. MCFA can cross the blood-brain barrier and in the brain can be oxidized through mitochondrial ß-oxidation. As components of ketogenic diets, MCFA have demonstrated beneficial effects on different brain diseases, such as traumatic brain injury, Alzheimer's disease, drug-resistant epilepsy, diabetes, and cancer. Despite the interest in MCFA effects, not much information is available about MCFA metabolism in the brain. In this study, with a gas chromatography-mass spectrometry (GC-MS)-based metabolomics approach, coupled with multivariate data analyses, we followed the metabolic changes of U87MG glioblastoma cells after the addition of octanoic (C8), or decanoic (C10) acids for 24 h. Our analysis highlighted significant differences in the metabolism of U87MG cells after the addition of C8 or C10 and identified several metabolites whose amount changed between the two groups of treated cells. Overall, metabolic pathway analyses suggested the citric acid cycle, Warburg effect, glutamine/glutamate metabolism, and ketone body metabolism as pathways influenced by C8 or C10 addition to U87MG cells. Our data demonstrated that, while C8 affected mitochondrial metabolism resulting in increased ketone body production, C10 mainly influenced cytosolic pathways by stimulating fatty acid synthesis. Moreover, glutamine might be the main substrate to support fatty acids synthesis in C10-treated cells. In conclusion, we identified a metabolic signature associated with C8 or C10 addition to U87MG cells that can be used to decipher metabolic responses of glioblastoma cells to MCFA treatment.

6.
Proteomes ; 8(3)2020 Aug 10.
Article in English | MEDLINE | ID: mdl-32784952

ABSTRACT

Ulcerative colitis (UC) and Crohn's disease (CD) represent the two main forms of chronic inflammatory bowel diseases (IBD). The exact IBD etiology is not yet revealed but CD and UC are likely induced by an excessive immune response against normal constituents of the intestinal microbial flora. IBD diagnosis is based on clinical symptoms often combined with invasive and costly procedures. Thus, the need for more non-invasive markers is urgent. Several routine laboratory investigations have been explored as indicators of intestinal inflammation in IBD, including blood testing for C-reactive protein, erythrocyte sedimentation rate, and specific antibodies, in addition to stool testing for calprotectin and lactoferrin. However, none has been universally adopted, some have been well-characterized, and others hold great promise. In recent years, the technological developments within the field of mass spectrometry (MS) and bioinformatics have greatly enhanced the ability to retrieve, characterize, and analyze large amounts of data. High-throughput research allowed enhancing the understanding of the biology of IBD permitting a more accurate biomarker discovery than ever before. In this review, we summarize currently used IBD serological and stool biomarkers and how proteomics and lipidomics are contributing to the identification of IBD biomarkers.

7.
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
9.
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
10.
Front Physiol ; 8: 544, 2017.
Article in English | MEDLINE | ID: mdl-28798698

ABSTRACT

ß-catenin plays an important role as regulatory hub in several cellular processes including cell adhesion, metabolism, and epithelial mesenchymal transition. This is mainly achieved by its dual role as structural component of cadherin-based adherens junctions, and as a key nuclear effector of the Wnt pathway. For this dual role, different classes of proteins are differentially regulated via ß-catenin dependent mechanisms. Here, we applied a liquid chromatography-mass spectrometry (LC-MS/MS) approach to identify proteins modulated after ß-catenin knockdown in the breast cancer cell line MCF-7. We used a label free analysis to compare trypsin-digested proteins from CTR (shCTR) and ß-catenin knockout cells (shßcat). This led to the identification of 98 differentially expressed proteins, 53 of them were up-regulated and 45 down-regulated. Loss of ß-catenin induced morphological changes and a significant modulation of the expression levels of proteins associated with primary metabolic processes. In detail, proteins involved in carbohydrate metabolism and tricarboxylic acid cycle were found to be down-regulated, whereas proteins associated to lipid metabolism were found up-regulated in shßcat compared to shCTR. A loss of mitochondrial mass and membrane potential was also assessed by fluorescent probes in shßcat cells with respect to the controls. These data are consistent with the reduced expression of transcriptional factors regulating mitochondrial biogenesis detected in shßcat cells. ß-catenin driven metabolic reprogramming resulted also in a significant modulation of lipogenic enzyme expression and activity. Compared to controls, ß-catenin knockout cells showed increased incorporation of [1-14C]acetate and decreased utilization of [U-14C]glucose for fatty acid synthesis. Our data highlight a role of ß-catenin in the regulation of metabolism and energy homeostasis in breast cancer cells.

11.
Mol Biosyst ; 13(6): 1131-1141, 2017 May 30.
Article in English | MEDLINE | ID: mdl-28429008

ABSTRACT

Resveratrol, a naturally occurring phytoalexin, has long been known to play an important regulatory role in key functions in cell physiology. This multifunctional role of resveratrol is explained by its ability to interact with several targets of various cell pathways. In the recent past, synthetic chemical modifications have been made in an attempt to enhance the biological effects of resveratrol, including its anti-cancer properties. In this study, we investigated the molecular mechanisms of action of novel trans-restricted analogues of resveratrol in which the C-C double bond of the natural derivative has been replaced by diaryl-substituted imidazole analogues. In ovarian cancer models, the results of in vitro screening revealed that the resveratrol analogues exhibited enhanced anti-proliferative properties compared with resveratrol. We found that the resveratrol analogues also significantly inhibited Akt and MAPK signalling and reduced the migration of IL-6 and EGF-treated cells. Finally, in ascite-derived cancer cells, we demonstrated that the resveratrol analogues reduced the expression of epithelial mesenchymal transition (EMT) markers. Collectively, these findings indicate the enhanced anti-cancer properties of the resveratrol analogues.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Ovarian Neoplasms/metabolism , Stilbenes/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Epidermal Growth Factor/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Interleukin-6/pharmacology , Resveratrol , Signal Transduction/drug effects , Stilbenes/chemistry
12.
J. physiol. biochem ; 72(3): 485-494, sept. 2016. tab, graf
Article in English | IBECS | ID: ibc-168290

ABSTRACT

The activities of lipogenic enzymes appear to fluctuate with changes in the level and type of dietary fats. Polyunsaturated fatty acids (PUFAs) are known to induce on hepatic de novo lipogenesis (DNL) the highest inhibitory effect, which occurs through a long-term adaptation. Data on the acute effects of dietary fatty acids on DNL are lacking. In this study with rats, the acute 1-day effect of high-fat (15 % w/w) diets (HFDs) enriched in saturated fatty acids (SFAs) or unsaturated fatty acids (UFAs), i.e., monounsaturated (MUFA) and PUFA, of the ω-6 and ω-3 series on DNL and plasma lipid level was investigated; a comparison with a longer time feeding (21 days) was routinely carried out. After 1-day HFD administration UFA, when compared to SFA, reduced plasma triacylglycerol (TAG) level and the activities of the lipogenic enzymes acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS), a decreased activity of the citrate carrier (CIC), a mitochondrial protein linked to lipogenesis, was also detected. In this respect, ω-3 PUFA was the most effective. On the other hand, PUFA maintained the effects at longer times, and the acute inhibition induced by MUFA feeding on DNL enzyme and CIC activities was almost nullified at 21 days. Mitochondrial fatty acid composition was slightly but significantly changed both at short- and long-term treatment, whereas the early changes in mitochondrial phospholipid composition vanished in long-term experiments. Our results suggest that in the early phase of administration, UFA coordinately reduced both the activities of de novo lipogenic enzymes and of CIC. ω-3 PUFA showed the greatest effect (AU)


No disponible


Subject(s)
Animals , Male , Diet, High-Fat/adverse effects , Lipids/blood , Lipogenesis , Liver/metabolism , Carrier Proteins/antagonists & inhibitors , Hypertriglyceridemia/prevention & control , Dietary Fats, Unsaturated/therapeutic use , Rats, Wistar , Time Factors , Triglycerides , Phospholipids/metabolism , Fatty Acids, Unsaturated , Fatty Acids, Monounsaturated , Mitochondria, Liver , Fatty Acid Synthases , Acetyl-CoA Carboxylase
13.
J Physiol Biochem ; 72(3): 485-94, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27312217

ABSTRACT

The activities of lipogenic enzymes appear to fluctuate with changes in the level and type of dietary fats. Polyunsaturated fatty acids (PUFAs) are known to induce on hepatic de novo lipogenesis (DNL) the highest inhibitory effect, which occurs through a long-term adaptation. Data on the acute effects of dietary fatty acids on DNL are lacking. In this study with rats, the acute 1-day effect of high-fat (15 % w/w) diets (HFDs) enriched in saturated fatty acids (SFAs) or unsaturated fatty acids (UFAs), i.e., monounsaturated (MUFA) and PUFA, of the ω-6 and ω-3 series on DNL and plasma lipid level was investigated; a comparison with a longer time feeding (21 days) was routinely carried out. After 1-day HFD administration UFA, when compared to SFA, reduced plasma triacylglycerol (TAG) level and the activities of the lipogenic enzymes acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS), a decreased activity of the citrate carrier (CIC), a mitochondrial protein linked to lipogenesis, was also detected. In this respect, ω-3 PUFA was the most effective. On the other hand, PUFA maintained the effects at longer times, and the acute inhibition induced by MUFA feeding on DNL enzyme and CIC activities was almost nullified at 21 days. Mitochondrial fatty acid composition was slightly but significantly changed both at short- and long-term treatment, whereas the early changes in mitochondrial phospholipid composition vanished in long-term experiments. Our results suggest that in the early phase of administration, UFA coordinately reduced both the activities of de novo lipogenic enzymes and of CIC. ω-3 PUFA showed the greatest effect.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Diet, High-Fat/adverse effects , Dietary Fats, Unsaturated/therapeutic use , Hypertriglyceridemia/prevention & control , Lipids/blood , Lipogenesis , Liver/metabolism , Acetyl-CoA Carboxylase/antagonists & inhibitors , Acetyl-CoA Carboxylase/metabolism , Animals , Carrier Proteins/metabolism , Dietary Fats, Unsaturated/adverse effects , Dietary Fats, Unsaturated/blood , Dietary Fats, Unsaturated/metabolism , Fatty Acid Synthases/antagonists & inhibitors , Fatty Acid Synthases/metabolism , Fatty Acids, Monounsaturated/adverse effects , Fatty Acids, Monounsaturated/blood , Fatty Acids, Monounsaturated/metabolism , Fatty Acids, Monounsaturated/therapeutic use , Fatty Acids, Omega-3/adverse effects , Fatty Acids, Omega-3/blood , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-3/therapeutic use , Fatty Acids, Omega-6/adverse effects , Fatty Acids, Omega-6/blood , Fatty Acids, Omega-6/metabolism , Fatty Acids, Omega-6/therapeutic use , Hypertriglyceridemia/blood , Hypertriglyceridemia/etiology , Hypertriglyceridemia/metabolism , Liver/enzymology , Male , Mitochondria, Liver/enzymology , Mitochondria, Liver/metabolism , Phospholipids/metabolism , Rats, Wistar , Time Factors , Triglycerides/antagonists & inhibitors , Triglycerides/blood , Triglycerides/metabolism
14.
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
15.
BMC Neurosci ; 16: 46, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-26205308

ABSTRACT

BACKGROUND: Lipids play different important roles in central nervous system so that dysregulation of lipid pathways has been implicated in a growing number of neurodegenerative disorders including multiple sclerosis (MS). MS is the most prevalent autoimmune disorder of the central nervous system, with neurological symptoms caused by inflammation and demyelination. In this study, a lipidomic analysis was performed for the rapid profile of CD4(+) T lymphocytes from MS patient and control samples in an untargeted approach. METHODS: A matrix-assisted laser desorption/ionization-time of flight (MALDI-TOF) mass spectrometry based approach was used for the analysis of lipid extracts using 9-aminoacridine as matrix. Lipids were analyzed in negative mode and selected species fragmented using MALDI tandem mass spectrometry for their structural assignments. RESULTS: The analysis reveals some modifications in the phospholipid pattern of MS CD4(+) T lymphocytes with respect to healthy controls with a significant increase of cardiolipin species in MS samples. CONCLUSIONS: These results demonstrate the feasibility of a MALDI-TOF approach for the analysis of CD4(+) lipid extracts and suggest how alterations in the lipid metabolism characterized lymphocytes of MS patients.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Cardiolipins/metabolism , Multiple Sclerosis, Relapsing-Remitting/metabolism , Phospholipids/metabolism , Adolescent , Adult , Discriminant Analysis , Fatty Acids/metabolism , Feasibility Studies , Female , Gas Chromatography-Mass Spectrometry , Humans , Least-Squares Analysis , Male , Middle Aged , Mitochondria/metabolism , Multivariate Analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Young Adult
16.
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
17.
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
18.
Biochim Biophys Acta ; 1771(10): 1299-307, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17905647

ABSTRACT

Metabolic fate and short-term effects of a 1:1 mixture of cis-9,trans-11 and trans-10,cis-12-conjugated linoleic acids (CLA), compared to linoleic acid (LA), on lipid metabolism was investigated in rat liver. In isolated mitochondria CLA-CoA were poorer substrates than LA-CoA for carnitine palmitoyltransferase-I (CPT-I) activity. However, in digitonin-permeabilized hepatocytes, where interactions among different metabolic pathways can be simultaneously investigated, CLA induced a remarkable stimulatory effect on CPT-I activity. This stimulation can be ascribed to a reduced malonyl-CoA level in turn due to inhibition of acetyl-CoA carboxylase (ACC) activity. The ACC/malonyl-CoA/CPT-I system can therefore represent a coordinate control by which CLA may exert effects on the partitioning of fatty acids between esterification and oxidation. Moreover, the rate of oxidation to CO2 and ketone bodies was significantly higher from CLA; peroxisomes rather than mitochondria were responsible for this difference. Interestingly, peroxisomal acyl-CoA oxidase (AOX) activity strongly increased by CLA-CoA compared to LA-CoA. CLA, metabolized by hepatocytes at a higher rate than LA, were poorer substrates for cellular and VLDL-triacylglycerol (TAG) synthesis. Overall, our results suggest that increased fatty acid oxidation with consequent decreased fatty acid availability for TAG synthesis is a potential mechanism by which CLA reduce TAG level in rat liver.


Subject(s)
Hepatocytes/metabolism , Linoleic Acids, Conjugated/metabolism , Liver/metabolism , Acetyl-CoA Carboxylase/metabolism , Acyl-CoA Oxidase , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Fatty Acids/metabolism , Lipids/chemistry , Mitochondria/metabolism , Models, Biological , Oxidoreductases/metabolism , Peroxisomes/metabolism , Rats , Rats, Wistar
19.
J Bioenerg Biomembr ; 39(2): 203-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17436146

ABSTRACT

All-trans-retinoic acid (atRA) is incorporated covalently into proteins of rat testes mitochondria. In this study, the effect of three diets with different fatty acid composition on the retinoylation of proteins of rat testes mitochondria has been investigated. Different groups of rats were fed on a basal diet supplemented with 15% of either coconut oil (CO), olive oil (OO) or fish oil (FO). We found that, when compared with CO, the binding of retinoic acid was decreased in FO- and OO-fed rats. Mitochondrial phospholipids composition was differently influenced by dietary treatments; minor changes were observed in fatty acid composition of phospholipids. Few differences were observed in the Arrhenius plots among the three groups of rats. Kinetic analysis revealed a decrease in the V (max) value in FO- and OO- as compared with CO-fed rats. No difference among the three groups were observed in the K ( M ) value. The retinoylation reaction was inhibited by 13-cis-RA and 9-cis-RA.


Subject(s)
Dietary Fats/metabolism , Mitochondria/metabolism , Phospholipids/metabolism , Testis/physiology , Tretinoin/metabolism , Animals , Coconut Oil , Fish Oils/metabolism , Male , Olive Oil , Plant Oils/metabolism , Rats , Rats, Wistar
20.
Biochim Biophys Acta ; 1761(4): 484-91, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16697699

ABSTRACT

The effect of hypothyroidism on citrate carrier (CiC) activity has been investigated in rat-liver mitochondria. The rate of citrate transport was reduced by approximately 50% in mitochondria from hypothyroid as compared with euthyroid rats. In parallel, a decrease in the rate of de novo fatty acid synthesis was observed in the cytosol of the former animals. Kinetic analysis of citrate transport revealed that only the Vmax was reduced by hypothyroidism, while Km was almost unaffected. Hypothyroidism increased the mitochondrial percentage of phosphatidylcholine while decreased that of phosphatidylethanolamine; an altered fatty acid pattern but no significant difference in the sum of saturated and unsaturated fatty acids as well as in the unsaturation index was observed. The CiC Arrhenius plot did not show appreciable difference between the two groups of rats. However, Western blot analysis associated with mRNA quantitation indicated that both protein level and mRNA accumulation of hepatic CiC were noticeably decreased in hypothyroid state. Therefore, a reduced content of the carrier protein can represent a plausible mechanism to explain the decline in the CiC activity observed in rat liver mitochondria of hypothyroid rats.


Subject(s)
Carrier Proteins/metabolism , Hypothyroidism/metabolism , Liver/metabolism , Mitochondria/metabolism , Acetyl-CoA Carboxylase/metabolism , Animals , Carrier Proteins/genetics , Citric Acid/metabolism , Down-Regulation , Fatty Acid Synthases/metabolism , Liver/cytology , Male , Membrane Lipids/chemistry , Mitochondria/chemistry , Rats , Rats, Wistar , Thyroid Hormones/blood
SELECTION OF CITATIONS
SEARCH DETAIL