Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 25(11)2024 May 30.
Article in English | MEDLINE | ID: mdl-38892185

ABSTRACT

N-methylpyridinium (NMP) is produced through the pyrolysis of trigonelline during the coffee bean roasting process. Preliminary studies suggest that NMP may have health benefits, thanks to its antioxidant properties. Based on this background, the aim of this study was to evaluate whether NMP could have a protective effect against LPS-induced neuroinflammation in human glioblastoma cells (U87MG). With this aim, U87MG cells were pre-treated with NMP (0.5 µM) for 1 h and then exposed to LPS (1 µg/mL) for 24 h. Our findings show that NMP attenuates LPS-induced neuroinflammation by reducing the expression of pro-inflammatory cytokines, such as IL-1ß, TNF-α and IL-6, through the inhibition of the NF-κB signaling pathway, which is critical in regulating inflammatory responses. NMP is able to suppress the activation of the NF-κB signaling pathway, suggesting its potential in preventing neuroinflammatory conditions. These outcomes support the notion that regular consumption of NMP, possibly through coffee consumption, may offer protection against neuroinflammatory states implicated in neurological disorders.


Subject(s)
Lipopolysaccharides , NF-kappa B , Neuroinflammatory Diseases , Neuroprotective Agents , Pyridinium Compounds , Signal Transduction , Humans , Neuroprotective Agents/pharmacology , NF-kappa B/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/chemically induced , Signal Transduction/drug effects , Pyridinium Compounds/pharmacology , Cell Line, Tumor , Cytokines/metabolism
2.
Int J Food Sci Nutr ; 74(4): 556-567, 2023.
Article in English | MEDLINE | ID: mdl-37431100

ABSTRACT

We investigated the extent to which adherence to the Mediterranean diet (MD) in combination with Mediterranean lifestyle factors influenced students' perceptions of subjective well-being (SWB) and distress. 939 undergraduates completed a survey to assess sociodemographic and lifestyle characteristics, including adherence to the MD, depression, anxiety, stress, and SWB. Data were analysed with correlation, logistic, and multiple linear regression models. Higher adherence to MD correlated with better SWB. Fruit, red meat, sweet and caffeinated beverages contributed significantly. However, it was the combination of adherence to MD with other factors, including quality of social relationships, income, smoking, sleep, and physical activity that better predicted SWB. Our results confirm the positive influence of MD on SWB. However, they also suggest the need to consider perceptions of well-being by a more holistic approach that considers physical and social factors simultaneously to improve the development of more effective educational and motivational programmes.


Subject(s)
Diet, Mediterranean , Life Style , Humans , Universities , Students , Italy , Perception
3.
J Bacteriol ; 204(2): e0046221, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34694905

ABSTRACT

The YjgF/YER057c/UK114 (Rid) is a protein family breadth conserved in all domains of life and includes the widely distributed archetypal RidA (YjgF) subfamily and seven other subfamilies (Rid1 to Rid7). Among these subfamilies, RidA is the only family to have been biochemically well characterized and is involved in the deamination of the reactive enamine/imine intermediates. In this study, we have characterized a protein of the Rid7 subfamily, named Rid7C, in Nonomuraea gerenzanensis, an actinomycete that is characterized by the presence of two types of RNA polymerases. This is due to the coexistence in its genome of two RNA polymerase (RNAP) ß chain-encoding genes, rpoB(S) (the wild-type rpoB gene) and rpoB(R) (a specialist, mutant-type rpoB gene) that controls A40926 antibiotic production and a wide range of metabolic adaptive behaviors. Here, we found that expression of rpoB(R) is regulated posttranscriptionally by RNA processing in the 5' untranslated region (UTR) of rpoB(R) mRNA and that the endoribonuclease activity of Rid7C is responsible for mRNA processing, thereby overseeing several tracts of morphological and biochemical differentiation. We also provide evidence that Rid7C may be associated with RNase P M1 RNA, although M1 RNA is not required for rpoB(R) mRNA processing in vitro, and that Rid7C endoribonuclease activity is inhibited by A40926, suggesting the existence of a negative feedback loop in A40926 production and a role of the endogenous synthesis of A40926 in the modulation of biochemical differentiation in this microorganism. IMPORTANCE The YjgF/YER057c/UK114 family includes many proteins with diverse functions involved in detoxification, RNA maturation, and control of mRNA translation. We found that Rid7C is an endoribonuclease that is involved in processing of rpoB(R) mRNA, coding for a specialized RNA polymerase beta subunit that oversees morphological differentiation and A40926 antibiotic production in Nonomuraea gerenzanensis. Rid7C-mediated processing promotes rpoB(R) mRNA translation and antibiotic production, while Rid7C endoribonuclease activity is inhibited by A40926, suggesting a role of the endogenous synthesis of A40926 in modulation of biochemical differentiation in this microorganism. Finally, we show that recombinant Rid7C copurified with M1 RNA (the RNA subunit of RNase P) from Escherichia coli extract, suggesting a functional interaction between Rid7C and M1 RNA activities.


Subject(s)
Actinobacteria/genetics , Actinobacteria/metabolism , DNA-Directed RNA Polymerases/genetics , Endoribonucleases/genetics , Gene Expression Regulation, Bacterial , Actinobacteria/drug effects , Actinobacteria/enzymology , Anti-Bacterial Agents/biosynthesis , Bacterial Proteins/metabolism , DNA-Directed RNA Polymerases/metabolism , Endoribonucleases/metabolism , Teicoplanin/analogs & derivatives , Teicoplanin/pharmacology
4.
Int J Mol Sci ; 23(3)2022 Jan 18.
Article in English | MEDLINE | ID: mdl-35162967

ABSTRACT

Dysregulation of de novo lipogenesis (DNL) has recently gained strong attention as being one of the critical factors that contribute to the assessment of non-alcoholic fatty liver disease (NAFLD). NAFLD is often diagnosed in patients with dyslipidemias and type 2 diabetes; thus, an interesting correlation can be deduced between high hematic free fatty acids and glucose excess in the DNL dysregulation. In the present study, we report that, in a cellular model of NAFLD, the coexistence of elevated glucose and FFA conditions caused the highest cellular lipid accumulation. Deepening the molecular mechanisms of the DNL dysregulation-RT-qPCR and immunoblot analysis demonstrated increased expression of mitochondrial citrate carrier (CiC), cytosolic acetyl-CoA carboxylase 1 (ACACA), and diacylglycerol acyltransferase 2 (DGAT2) involved in fatty acids and triglycerides synthesis, respectively. XBP-1, an endoplasmic reticulum stress marker, and SREBP-1 were the transcription factors connected to the DNL activation. Quercetin (Que), a flavonoid with strong antioxidant properties, and noticeably reduced the lipid accumulation and the expression of SREBP-1 and XBP-1, as well as of their lipogenic gene targets in steatotic cells. The anti-lipogenic action of Que mainly occurs through a strong phosphorylation of ACACA, which catalyzes the committing step in the DNL pathway. The high level of ACACA phosphorylation in Que-treated cells was explained by the intervention of AMPK together with the reduction of enzymatic activity of PP2A phosphatase. Overall, our findings highlight a direct anti-lipogenic effect of Que exerted through inhibition of the DNL pathway by acting on ACACA/AMPK/PP2A axis; thus, suggesting this flavonoid as a promising molecule for the NAFLD treatment.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Fatty Acids/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Quercetin/pharmacology , AMP-Activated Protein Kinases/metabolism , Acetyl-CoA Carboxylase/genetics , Gene Expression Profiling , Hep G2 Cells , Humans , Lipogenesis , Models, Biological , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/genetics , Phosphorylation , Protein Phosphatase 2/metabolism , Signal Transduction
5.
Int J Mol Sci ; 23(23)2022 Dec 02.
Article in English | MEDLINE | ID: mdl-36499489

ABSTRACT

Edentulism is the condition of having lost natural teeth, and has serious social, psychological, and emotional consequences. The need for implant services in edentulous patients has dramatically increased during the last decades. In this study, the effects of concentrated growth factor (CGF), an autologous blood-derived biomaterial, in improving the process of osseointegration of dental implants have been evaluated. Here, permeation of dental implants with CGF has been obtained by using a Round up device. These CGF-coated dental implants retained a complex internal structure capable of releasing growth factors (VEGF, TGF-ß1, and BMP-2) and matrix metalloproteinases (MMP-2 and MMP-9) over time. The CGF-permeated implants induced the osteogenic differentiation of human bone marrow stem cells (hBMSC) as confirmed by matrix mineralization and the expression of osteogenic differentiation markers. Moreover, CGF provided dental implants with a biocompatible and biologically active surface that significantly improved adhesion of endothelial cells on CGF-coated implants compared to control implants (without CGF). Finally, data obtained from surgical interventions with CGF-permeated dental implants presented better results in terms of optimal osseointegration and reduced post-surgical complications. These data, taken together, highlight new and interesting perspectives in the use of CGF in the dental implantology field to improve osseointegration and promote the healing process.


Subject(s)
Dental Implants , Osteogenesis , Humans , Endothelial Cells , Intercellular Signaling Peptides and Proteins/pharmacology , Osseointegration , Surface Properties , Titanium/pharmacology , Coated Materials, Biocompatible/pharmacology
6.
Int J Mol Sci ; 22(8)2021 Apr 07.
Article in English | MEDLINE | ID: mdl-33916919

ABSTRACT

Reactive intermediate deaminase (Rid) proteins are enzymes conserved in all domains of life. UK114, a mammalian member of RidA subfamily, has been firstly identified as a component of liver perchloric acid-soluble proteins (L-PSP). Although still poorly defined, several functions have been attributed to the mammalian protein UK114/RIDA, including the reactive intermediate deamination activity. The expression of UK114/RIDA has been observed in some tumors, arousing interest in this protein as an evaluable tumor marker. However, other studies reported a negative correlation between UK114/RIDA expression, tumor differentiation degree and cell proliferation. This work addressed the question of UK114/RIDA expression in human non-tumor HEK293 cell lines and in some human tumor cell lines. Here we reported that human RIDA (hRIDA) was expressed in all the analyzed cell line and subjected to lysine (K-)succinylation. In HEK293, hRIDA K-succinylation was negatively correlated to the cell proliferation rate and was under the control of SIRT5. Moreover, K-succinylation clearly altered hRIDA quantification by immunoblotting, explaining, at least in part, some discrepancies about RIDA expression reported in previous studies. We found that hRIDA was able to deaminate reactive enamine-imine intermediates and that K-succinylation drastically reduced deaminase activity. As predicted by in silico analysis, the observed reduction of deaminase activity has been related to the drastic alterations of hRIDA structure inferred by K-succinylation. The role of hRIDA and the importance of its K-succinylation in cell metabolism, especially in cancer biology, have been discussed.


Subject(s)
Cell Proliferation/physiology , Heat-Shock Proteins/metabolism , Ribonucleases/metabolism , Cell Line , Enzyme Activation , Gene Expression , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/genetics , Humans , Lysine/metabolism , Models, Molecular , Protein Conformation , Protein Processing, Post-Translational , Ribonucleases/chemistry , Ribonucleases/genetics , Sirtuins/metabolism , Structure-Activity Relationship
7.
Int J Mol Sci ; 22(16)2021 Aug 18.
Article in English | MEDLINE | ID: mdl-34445573

ABSTRACT

Concentrated Growth Factors (CGF) represent new autologous (blood-derived biomaterial), attracting growing interest in the field of regenerative medicine. In this study, the chemical, structural, and biological characterization of CGF was carried out. CGF molecular characterization was performed by GC/MS to quantify small metabolites and by ELISA to measure growth factors and matrix metalloproteinases (MMPs) release; structural CGF characterization was carried out by SEM analysis and immunohistochemistry; CGF has been cultured, and its primary cells were isolated for the identification of their surface markers by flow cytometry, Western blot, and real-time PCR; finally, the osteogenic differentiation of CGF primary cells was evaluated through matrix mineralization by alizarin red staining and through mRNA quantification of osteogenic differentiation markers by real-time PCR. We found that CGF has a complex inner structure capable of influencing the release of growth factors, metabolites, and cells. These cells, which could regulate the production and release of the CGF growth factors, show stem features and are able to differentiate into osteoblasts producing a mineralized matrix. These data, taken together, highlight interesting new perspectives for the use of CGF in regenerative medicine.


Subject(s)
Cell Differentiation , Intercellular Signaling Peptides and Proteins/pharmacology , Osteoblasts/cytology , Osteogenesis , Stem Cells/cytology , Cell Proliferation , Cells, Cultured , Female , Humans , Male , Osteoblasts/drug effects , Osteoblasts/metabolism , Stem Cells/drug effects , Stem Cells/metabolism
8.
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
9.
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
10.
IUBMB Life ; 71(7): 863-872, 2019 07.
Article in English | MEDLINE | ID: mdl-30707786

ABSTRACT

Hepatic de novo lipogenesis (DNL), the process by which carbohydrates are converted into lipids, is strictly controlled by nutritional and hormonal status. 3,5-Diiodo-L-thyronine (T2), a product of the 3,5,3'-triiodo-L-thyronine (T3) peripheral metabolism, has been shown to mimic some T3 effects on lipid metabolism by a short-term mechanism independent of protein synthesis. Here, we report that T2, administered for 1 week to hypothyroid rats, increases total fatty acid synthesis from acetate in isolated hepatocytes. Studies carried out on liver subcellular fractions demonstrated that T2 not only increases the activity and the expression of acetyl-CoA carboxylase and fatty acid synthase but also of other proteins linked to DNL such as the mitochondrial citrate carrier and the cytosolic ATP citrate lyase. Parallelly, T2 stimulates the activities of enzymes supplying cytosolic NADPH needed for the reductive steps of DNL. With respect to both euthyroid and hypothyroid rats, T2 administration decreases the hepatic mRNA level of SREBP-1, a transcription factor which represents a master regulator of DNL. However, when compared to hypothyroid rats T2 significantly increases, without bringing to the euthyroid value, the content of both mature (nSREBP-1), and precursor (pSREBP-1) forms of the SREBP-1 protein as well as their ratio. Moreover, T2 administration strongly augmented the nuclear content of ChREBP, another crucial transcription factor involved in the regulation of lipogenic genes. Based on these results, we can conclude that in the liver of hypothyroid rats the transcriptional activation by T2 of DNL genes could depend, at least in part, on SREBP-1- and ChREBP-dependent mechanisms. © 2019 IUBMB Life, 2019.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Diiodothyronines/pharmacology , Gene Expression Regulation/drug effects , Hypothyroidism/metabolism , Lipogenesis/drug effects , Liver/physiology , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hypothyroidism/drug therapy , Hypothyroidism/pathology , Liver/drug effects , Male , Rats , Rats, Wistar , Sterol Regulatory Element Binding Protein 1/genetics , Transcriptional Activation
11.
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
12.
Metab Eng ; 48: 254-268, 2018 07.
Article in English | MEDLINE | ID: mdl-29944936

ABSTRACT

Pirins are evolutionarily conserved iron-containing proteins that are found in all kingdoms of life, and have been implicated in diverse molecular processes, mostly associated with cellular stress. In the present study, we started from the evidence that the insertional inactivation of pirin-like gene SAM23877_RS18305 (pirA) by ΦC31 Att/Int system-based vectors in spiramycin-producing strain Streptomyces ambofaciens ATCC 23877 resulted in marked effects on central carbon and energy metabolism gene expression, high sensitivity to oxidative injury and repression of polyketide antibiotic production. By using integrated transcriptomic, proteomic and metabolite profiling, together with genetic complementation, we here show that most of these effects could be traced to the inability of the pirA-defective strain to modulate beta-oxidation pathway, leading to an unbalanced supply of precursor monomers for polyketide biosynthesis. Indeed, in silico protein-protein interaction modeling and in vitro experimental validation allowed us to demonstrate that PirA is a novel redox-sensitive negative modulator of very long-chain acyl-CoA dehydrogenase, which catalyzes the first committed step of the beta-oxidation pathway.


Subject(s)
Bacterial Proteins , Iron-Binding Proteins , Metabolic Engineering , Streptomyces , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Oxidation-Reduction , Polyketides/metabolism , Streptomyces/genetics , Streptomyces/metabolism
13.
J Biol Chem ; 291(38): 19746-59, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27476175

ABSTRACT

Heme is an essential molecule in many biological processes, such as transport and storage of oxygen and electron transfer as well as a structural component of hemoproteins. Defects of heme biosynthesis in developing erythroblasts have profound medical implications, as represented by sideroblastic anemia. The synthesis of heme requires the uptake of glycine into the mitochondrial matrix where glycine is condensed with succinyl coenzyme A to yield δ-aminolevulinic acid. Herein we describe the biochemical and molecular characterization of yeast Hem25p and human SLC25A38, providing evidence that they are mitochondrial carriers for glycine. In particular, the hem25Δ mutant manifests a defect in the biosynthesis of δ-aminolevulinic acid and displays reduced levels of downstream heme and mitochondrial cytochromes. The observed defects are rescued by complementation with yeast HEM25 or human SLC25A38 genes. Our results identify new proteins in the heme biosynthetic pathway and demonstrate that Hem25p and its human orthologue SLC25A38 are the main mitochondrial glycine transporters required for heme synthesis, providing definitive evidence of their previously proposed glycine transport function. Furthermore, our work may suggest new therapeutic approaches for the treatment of congenital sideroblastic anemia.


Subject(s)
Anemia/metabolism , Heme/biosynthesis , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Anemia/genetics , Genetic Complementation Test , Glycine/genetics , Glycine/metabolism , Heme/genetics , Humans , Mitochondria/genetics , Mitochondrial Membrane Transport Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics
14.
Biochim Biophys Acta ; 1861(5): 471-81, 2016 May.
Article in English | MEDLINE | ID: mdl-26869449

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a chronic disease characterized by accumulation of lipid droplets in hepatocytes. Enhanced release of non-esterified fatty acids from adipose tissue accounts for a remarkable fraction of accumulated lipids. However, the de novo lipogenesis (DNL) is also implicated in the etiology of the NAFLD. Sterol Regulatory Element-Binding Protein-1 (SREBP-1) is a transcription factor modulating the expression of several lipogenic enzymes. In the present study, in order to investigate the effect of lipid droplet accumulation on DNL, we used a cellular model of steatosis represented by HepG2 cells cultured in a medium supplemented with free oleic and palmitic fatty acids (FFAs). We report that FFA supplementation induces the expression of genes coding for enzymes involved in the DNL as well as for the transcription factor SREBP-1a. The SREBP-1a mRNA translation, dependent on an internal ribosome entry site (IRES), and the SREBP-1a proteolytic cleavage are activated by FFAs. Furthermore, FFA treatment enhances the expression and the nucleus-cytosolic shuttling of hnRNP A1, a trans-activating factor of SREBP-1a IRES. The binding of hnRNP A1 to the SREBP-1a IRES is also increased upon FFA supplementation. The relocation of hnRNP A1 and the consequent increase of SREBP-1a translation are dependent on the p38 MAPK signal pathway, which is activated by FFAs. By RNA interference approach, we demonstrate that hnRNP A1 is implicated in the FFA-induced expression of SREBP-1a and of its target genes as well as in the lipid accumulation in cells.


Subject(s)
5' Untranslated Regions , Hepatocytes/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Lipogenesis , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , RNA, Messenger/metabolism , Sterol Regulatory Element Binding Protein 1/biosynthesis , Binding Sites , Gene Expression Regulation , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/pathology , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Humans , Internal Ribosome Entry Sites , Lipogenesis/drug effects , Lipogenesis/genetics , Liver/drug effects , Liver/pathology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Oleic Acid/pharmacology , Palmitic Acid/pharmacology , Protein Transport , RNA Interference , RNA, Messenger/genetics , Signal Transduction , Sterol Regulatory Element Binding Protein 1/genetics , Time Factors , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Int J Mol Sci ; 18(4)2017 Mar 31.
Article in English | MEDLINE | ID: mdl-28362337

ABSTRACT

The thyroid hormones (THs) 3,3',5,5'-tetraiodo-l-thyronine (T4) and 3,5,3'-triiodo-l-thyronine (T3) influence many metabolic pathways. The major physiological function of THs is to sustain basal energy expenditure, by acting primarily on carbohydrate and lipid catabolism. Beyond the mobilization and degradation of lipids, at the hepatic level THs stimulate the de novo fatty acid synthesis (de novo lipogenesis, DNL), through both the modulation of gene expression and the rapid activation of cell signalling pathways. 3,5-Diiodo-l-thyronine (T2), previously considered only a T3 catabolite, has been shown to mimic some of T3 effects on lipid catabolism. However, T2 action is more rapid than that of T3, and seems to be independent of protein synthesis. An inhibitory effect on DNL has been documented for T2. Here, we give an overview of the mechanisms of THs action on liver fatty acid metabolism, focusing on the different effects exerted by T2 and T3 on the regulation of the DNL. The inhibitory action on DNL exerted by T2 makes this compound a potential and attractive drug for the treatment of some metabolic diseases and cancer.


Subject(s)
Diiodothyronines/metabolism , Fatty Acids/metabolism , Liver/metabolism , Thyroxine/metabolism , Triiodothyronine/metabolism , Animals , Humans , Lipid Metabolism , Mitochondria/metabolism , Models, Biological
16.
Biochim Biophys Acta ; 1849(1): 23-31, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25450523

ABSTRACT

The Unfolded Protein Response (UPR) is an intracellular signaling pathway which is activated when unfolded or misfolded proteins accumulate in the Endoplasmic Reticulum (ER), a condition commonly referred to as ER stress. It has been shown that lipid biosynthesis is increased in ER-stressed cells. The N(ε)-lysine acetylation of ER-resident proteins, including chaperones and enzymes involved in the post-translational protein modification and folding, occurs upon UPR activation. In both ER proteins acetylation and lipid synthesis, acetyl-CoA is the donor of acetyl group and it is transported from the cytosol into the ER. The cytosolic pool of acetyl-CoA is mainly derived from the activity of mitochondrial citrate carrier (CiC). Here, we have demonstrated that expression of CiC is activated in human HepG2 and rat BRL-3A cells during tunicamycin-induced ER stress. This occurs through the involvement of an ER stress responsive region identified within the human and rat CiC proximal promoter. A functional Unfolded Protein Response Element (UPRE) confers responsiveness to the promoter activation by UPR transducers ATF6α and XBP1. Overall, our data demonstrate that CiC expression is activated during ER stress through the binding of ATF6α and XBP1 to an UPRE element located in the proximal promoter of Cic gene. The role of ER stress-mediated induction of CiC expression has been discussed.


Subject(s)
Activating Transcription Factor 6/genetics , Carrier Proteins/metabolism , DNA-Binding Proteins/genetics , Signal Transduction , Transcription Factors/genetics , Unfolded Protein Response/genetics , Acetyl Coenzyme A/metabolism , Activating Transcription Factor 6/metabolism , Animals , Carrier Proteins/genetics , Citric Acid/metabolism , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/genetics , Hep G2 Cells , Humans , Promoter Regions, Genetic , Protein Folding , Protein Processing, Post-Translational , Rats , Regulatory Factor X Transcription Factors , X-Box Binding Protein 1
17.
J Bioenerg Biomembr ; 48(5): 521-529, 2016 10.
Article in English | MEDLINE | ID: mdl-27854029

ABSTRACT

The role of 3,5-diiodo-L-thyronine (T2), initially considered only a 3,3',5-triiodo-L-thyronine (T3) catabolite, in the bioenergetic metabolism is of growing interest. In this study we investigated the acute effects (within 1 h) of T2 administration to hypothyroid rats on liver mitochondria fatty acid uptake and ß-oxidation rate, mitochondrial efficiency (by measuring proton leak) and mitochondrial oxidative damage (by determining H2O2 release). Fatty acid uptake into mitochondria was measured assaying carnitine palmitoyl transferase (CPT) I and II activities, and fatty acid ß-oxidation using palmitoyl-CoA as a respiratory substrate. Mitochondrial fatty acid pattern was defined by gas-liquid chromatography. In hypothyroid + T2 vs hypothyroid rats we observed a raise in the serum level of nonesterified fatty acids (NEFA), in the mitochondrial CPT system activity and in the fatty acid ß-oxidation rate. A parallel increase in the respiratory chain activity, mainly from succinate, occurs. When fatty acids are chelated by bovine serum albumin, a T2-induced increase in both state 3 and state 4 respiration is observed, while, when fatty acids are present, mitochondrial uncoupling occurs together with increased proton leak, responsible for mitochondrial thermogenesis. T2 administration decreases mitochondrial oxidative stress as determined by lower H2O2 production. We conclude that in rat liver mitochondria T2 acutely enhances the rate of fatty acid ß-oxidation, and the activity of the downstream respiratory chain. The T2-induced increase in proton leak may contribute to mitochondrial thermogenesis and to the reduction of oxidative stress. Our results strengthen the previously reported ability of T2 to reduce adiposity, dyslipidemia and to prevent liver steatosis.


Subject(s)
Diiodothyronines/pharmacology , Energy Metabolism/drug effects , Hypothyroidism/drug therapy , Mitochondria, Liver/metabolism , Animals , Diiodothyronines/administration & dosage , Electron Transport/drug effects , Fatty Acids/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Rats , Uncoupling Agents/pharmacology
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL