Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Nutr Metab Cardiovasc Dis ; 31(12): 3522-3532, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34629256

ABSTRACT

BACKGROUND AND AIMS: Several chronic multifactorial diseases originate from energy unbalance between food intake and body energy expenditure, including non-alcoholic fatty liver disease (NAFLD), diabetes, and cardiovascular disorders. Vascular endothelium plays a central role in body homeostasis, and NAFLD is often associated with endothelial dysfunction (ED), the first step in atherosclerosis. Both sugars and fatty acids (FAs) are fuel sources for energy production, but their excess leads to liver steatosis which may trigger ED through a network of mechanisms which need to be clarified. Here, we investigated the crosstalk pathways between in vitro cultured steatotic hepatocytes (FaO) and endothelial cells (HECV) being mediated by soluble factors. METHODS AND RESULTS: We employed the conditioned medium approach to test how different extent and features of hepatic steatosis distinctively affect endothelium leading to ED. The steatogenic media collected from steatotic hepatocytes were characterized by high triglyceride content and led to lipid accumulation and fat-dependent dysfunction in HECV cells. We found a parallelism between (i) extent of hepatocyte steatosis and level of lipid accumulation in HECV cells; (ii) type of hepatocyte steatosis (with macro- or microvesicular LDs) and extent of oxidative stress, lipid peroxidation, nitric oxide release and expression of ED markers in HECV cells. CONCLUSIONS: The present findings seem to suggest that, in addition to triglycerides, other soluble mediators should be released by steatotic hepatocytes and may influence lipid accumulation and function of HECV cells. Further studies need to depict the exact profile of soluble factors involved in steatotic hepatocyte-endothelium crosstalk.


Subject(s)
Endothelial Cells , Fatty Liver , Cell Communication , Endothelial Cells/physiology , Fatty Liver/physiopathology , Humans
2.
Life Sci ; 265: 118812, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33278396

ABSTRACT

AIMS: Adipocyte hypertrophy is the main cause of obesity. A deeper understanding of the molecular mechanisms regulating adipocyte dysfunction may help to plan strategies to treat/prevent obesity and its metabolic complications. Here, we investigated in vitro the molecular alterations associated with early adipocyte hypertrophy, focusing on mitochondrial dysfunction. MAIN METHODS: As model of adipocyte hypertrophy, we employed 3T3-L1 preadipocytes firstly differentiated into mature adipocytes, then cultured with long-chain fatty acids. As a function of differentiation and hypertrophy, we assessed triglyceride content, lipid droplet size, radical homeostasis by spectrophotometry and microscopy, as well as the expression of PPARγ, adiponectin and metallothioneins. Mitochondrial status was investigated by electron microscopy, oxygraph 2 k (O2K) high-resolution respirometry, fluorimetry and western blot. KEY FINDINGS: Compared to mature adipocytes, hypertrophic adipocytes showed increased triglyceride accumulation and lipid peroxidation, larger or unique lipid droplet, up-regulated expression of PPARγ, adiponectin and metallothioneins. At mitochondrial level, early-hypertrophic adipocytes exhibited: (i) impaired mitochondrial oxygen consumption with parallel reduction in the mitochondrial complexes; (ii) no changes in citrate synthase and HSP60 expression, and in the inner mitochondrial membrane polarization; (iii) no stimulation of mitochondrial fatty acid oxidation. Our findings indicate that the content, integrity, and catabolic activity of mitochondria were rather unchanged in early hypertrophic adipocytes, while oxygen consumption and oxidant production were altered. SIGNIFICANCE: In the model of early adipocyte hypertrophy exacerbated oxidative stress and impaired mitochondrial respiration were observed, likely depending on reduction in the mitochondrial complexes, without changes in mitochondrial mass and integrity.


Subject(s)
Adipocytes/pathology , Adipose Tissue/pathology , Mitochondria/pathology , Obesity/physiopathology , 3T3-L1 Cells , Animals , Cell Differentiation , Electron Transport/physiology , Hypertrophy , Mice , Oxidative Stress/physiology , Oxygen Consumption/physiology
3.
Nutr Cancer ; 73(7): 1193-1206, 2021.
Article in English | MEDLINE | ID: mdl-32696667

ABSTRACT

Thyme-like plants including Thymbra spicata L. are widely used as food and folk medicinal remedies in the Mediterranean area. This study aimed to explore the in vitro antitumor potential of polyphenol-enriched extracts from aerial parts of T. spicata. The ethanolic extract significantly inhibited proliferation of different human tumor cell lines, without significant effects on non-neoplastic cells. A deeper investigation of the molecular mechanism sustaining the in vitro antitumor activity of the extract was carried on the human breast cancer cells MCF-7 in comparison with the normal breast cells MCF-10A. The effects on MCF-7 cells were associated with the following: (i) production of reactive oxygen species (ROS) and release of nitric oxide; (ii) apoptosis induction; and (iii) reduction in STAT3 and NF-kB phosphorylation. The ethanolic extract from T. spicata leaves might represent a novel therapeutic tool in combination with conventional chemotherapy to reduce the adverse side effects and drug resistance.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Lamiaceae/chemistry , NF-kappa B , Plant Extracts , Apoptosis , Cell Proliferation , Cell Survival , Humans , MCF-7 Cells , Plant Extracts/pharmacology , Plant Leaves/chemistry , STAT3 Transcription Factor
4.
Molecules ; 25(18)2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32942773

ABSTRACT

S-adenosylmethionine (SAMe) is an endogenous methyl donor derived from ATP and methionine that has pleiotropic functions. Most SAMe is synthetized and consumed in the liver, where it acts as the main methylating agent and in protection against the free radical toxicity. Previous studies have shown that the administration of SAMe as a supernutrient exerted many beneficial effects in various tissues, mainly in the liver. In the present study, we aimed to clarify the direct effects of SAMe on fatty acid-induced steatosis and oxidative stress in hepatic and endothelial cells. Hepatoma FaO cells and endothelial HECV cells exposed to a mixture of oleate/palmitate are reliable models for hepatic steatosis and endothelium dysfunction, respectively. Our findings indicate that SAMe was able to significantly ameliorate lipid accumulation and oxidative stress in hepatic cells, mainly through promoting mitochondrial fatty acid entry for ß-oxidation and external triglyceride release. SAMe also reverted both lipid accumulation and oxidant production (i.e., ROS and NO) in endothelial cells. In conclusion, these outcomes suggest promising beneficial applications of SAMe as a nutraceutical for metabolic disorders occurring in fatty liver and endothelium dysfunction.


Subject(s)
Oxidative Stress/drug effects , S-Adenosylmethionine/pharmacology , Animals , Cell Line, Tumor , Cell Movement/drug effects , Endothelial Cells/cytology , Endothelial Cells/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Malondialdehyde/metabolism , Nitric Oxide/metabolism , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/drug therapy , Oleanolic Acid/toxicity , Palmitic Acid/toxicity , Rats , Reactive Oxygen Species/metabolism , S-Adenosylmethionine/therapeutic use
5.
Article in English | MEDLINE | ID: mdl-31816412

ABSTRACT

Hepatic steatosis is the hallmark of non-alcoholic fatty liver disease (NAFLD), the hepatic manifestation of the metabolic syndrome and insulin resistance with potential evolution towards non-alcoholic steatohepatitis (NASH), cirrhosis and hepatocellular carcinoma. Key roles of autophagy and oxidative stress in hepatic lipid accumulation and NAFLD progression are recognized. Here, we employed a rat hepatoma cell model of NAFLD progression made of FaO cells exposed to oleate/palmitate followed or not by TNFα treatment to investigate the molecular mechanisms through which silybin, a lipid-lowering nutraceutical, may improve hepatic lipid dyshomeostasis. The beneficial effect of silybin was found to involve amelioration of the fatty acids profile of lipid droplets, stimulation of the mitochondrial oxidation and upregulation of a microRNA of pivotal relevance in hepatic fat metabolism, miR-122. Silybin was also found to restore the levels of Aquaporin-9 (AQP9) and glycerol permeability while reducing the activation of the oxidative stress-dependent transcription factor NF-κB, and autophagy turnover. In conclusion, silybin was shown to have molecular effects on signaling pathways that were previously unknown and potentially protect the hepatocyte. These actions intersect TG metabolism, fat-induced autophagy and AQP9-mediated glycerol transport in hepatocytes.


Subject(s)
Aquaporins/metabolism , Autophagy , Hepatocytes/metabolism , Lipid Droplets/metabolism , Lipid Metabolism , Silybin/pharmacology , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Animals , Aquaporins/genetics , Cell Line, Tumor , Hepatocytes/drug effects , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Rats
6.
J Mech Behav Biomed Mater ; 97: 296-305, 2019 09.
Article in English | MEDLINE | ID: mdl-31151002

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a chronic liver disease often associated with overnutrition. Number and morphometry of lipid droplets (LDs) define micro vs macrovesicular steatosis, influence the morphology and function of hepatocytes and possibly their stiffness. The link between grade and features of steatosis and biomechanical properties of single hepatocytes requires deeper investigations. In vitro NAFLD models with distinct steatosis conditions were set by exposing FaO hepatoma cells to single or combined fructose (Fru), fatty acids (FA), and tumor necrosis factor (TNF)α. Single Cell Force Spectroscopy and Quantitative Phase Microscopy quantified the single cell stiffness and a series of morphometric parameters; the mRNA expression of genes involved in lipid metabolism was quantified by real-time PCR. In our models, LD size and number increased with Fru and FA as single agents, and more with combined Fru/FA (macrovesicular steatosis), while FA/TNFα combination increased LD number with a reduction in their size (microvesicular steatosis). We found that the changes in LD size and number influenced cell stiffness and morphometry as follows: (i) single cell elasticity increased in macrovesicular steatosis (maximally with combined Fru/FA); (ii) FA-induced steatosis resulted in cells thinner and larger, whereas combined FA/TNFα shrunk the hepatocytes. Taken together the data on hepatocyte biomechanics show that, in addition to extent of lipid accumulation, cell stiffness is mainly influenced by LD size, while cell morphometry directly relates to LD number. Our findings suggest that a novel mechanobiology perspective might provide future contributions in NAFLD research.


Subject(s)
Carcinoma, Hepatocellular/pathology , Fatty Liver/pathology , Hepatocytes/cytology , Lipid Droplets/chemistry , Liver Neoplasms/pathology , Non-alcoholic Fatty Liver Disease/pathology , Animals , Biomarkers/metabolism , Biomechanical Phenomena , Biophysics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Survival , Elasticity , Fatty Acids/metabolism , Fructose/chemistry , Lipids/chemistry , Liver/cytology , Liver Neoplasms/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Rats , Tumor Necrosis Factor-alpha/metabolism
7.
Int J Mol Med ; 44(2): 705-712, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31173180

ABSTRACT

Overconsumption of fats and sugars is a major cause of development of non­alcoholic fatty liver disease (NAFLD). The main objectives of the present study were to explore the pathways sustaining the interfering metabolic effects of excess fructose and fatty acids in hepatocytes, and to clarify the mechanisms through which the nutraceutical silybin rescues the functional and metabolic alterations that are associated with the NALFD progression. Cultured hepatocytes were exposed to fructose and fatty acids, alone or in combination, to induce different grades of steatosis in vitro. Cell viability, apoptosis, free radical production, lipid content, lipid peroxidation and activity of lipogenic enzymes were assessed by spectrophotometric assays. Oxygen consumption and mitochondrial respiration parameters were measured using a Seahorse analyzer. Expression of markers for liver steatosis and dysfunction were also evaluated by reverse transcription­quantitative polymerase chain reaction. The data revealed that fructose and fatty acid combination in vitro had a positive interference on lipogenic pathways, leading to more severe steatosis and liver dysfunction, reduced cell viability, increased apoptosis, oxidative stress and mitochondrial respiration. Hepatic cell abnormalities were almost completely alleviated by silybin treatment. These findings suggest that silybin may serve as a novel and cost­effective dietary supplement for treatment and/or prevention of hepatosteatosis associated with NAFLD progression.


Subject(s)
Fatty Acids/metabolism , Fructose/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Protective Agents/pharmacology , Silybin/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Lipid Metabolism/drug effects , Lipid Peroxidation/drug effects , Non-alcoholic Fatty Liver Disease/metabolism , Oxidative Stress/drug effects , Rats
8.
J Ethnopharmacol ; 239: 111919, 2019 Jul 15.
Article in English | MEDLINE | ID: mdl-31029756

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Thymbra spicata, a member of the Lamiaceae family, is native to eastern Mediterranean area. Leaves of this plant are rich in phenolic compounds and are a popular remedy of traditional medicine in Lebanon to prevent and/or counteract hyperlipidemia and hyperglycemia. AIM OF THE STUDY: To evaluate the antisteatotic and antioxidant activities of extracts from leaves of Thymbra spicata L. using in vitro models of non-alcoholic fatty liver disease (NAFLD), a leading cause of liver-related morbidity and mortality worldwide, for whom no effective treatments are still available. MATERIALS AND METHODS: Two different extracts from Thymbra spicata L. aerial parts were prepared using water (TW) or ethanol (TE) as solvent. Their chemical composition was characterized by gas and liquid chromatography coupled with mass spectrometry. Both extracts were tested on cultured hepatic and endothelial cells treated to mimic in vitro a multisistemic pathology such as NAFLD. We assayed the effects on lipid accumulation, free radical production, lipid peroxidation, cell migration. RESULTS: Both the total phenolic and the total flavonoid contents were higher in the ethanolic extract. Rosmarinic acid was the most abundant polyphenol in TW, while TE was richer in carvacrol. Our findings demonstrated that both extracts ameliorated lipid accumulation, oxidative stress and inflammation in the NAFLD cellular models. However, the aqueous extract was more effective to reduce hepatic steatosis, and the ethanolic extract had higheranti-oxidant potential and wound healing activity. CONCLUSIONS: T. spicata extracts could be promising bioactive products to develop natural therapeutic agents or dietary supplements to treat NAFLD and obesity-related metabolic disease. Our findings suggest that while the ethanolic extract might be used in preventing endothelium dysfunction, the aqueous extract would act better as lipid-lowering agent.


Subject(s)
Antioxidants/pharmacology , Endothelial Cells/drug effects , Lamiaceae , Non-alcoholic Fatty Liver Disease/drug therapy , Plant Extracts/pharmacology , Animals , Cell Movement/drug effects , Cells, Cultured , Endothelial Cells/physiology , Humans , Lipid Metabolism/drug effects , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Plant Components, Aerial , Rats , Transcription Factor RelA/metabolism , Wound Healing/drug effects
9.
Eur J Nutr ; 57(5): 1793-1805, 2018 Aug.
Article in English | MEDLINE | ID: mdl-28526925

ABSTRACT

PURPOSE: Phenolic compounds (PC) of virgin olive oil exert several biochemical and pharmacological beneficial effects. Some dietary PC seem to prevent/improve obesity and metabolic-related disorders such as non-alcoholic fatty liver disease (NAFLD). We investigated the possible effects of PC extracted from olive pomace (PEOP) and of the main single molecules present in the extract (tyrosol, apigenin, oleuropein, p-coumaric and caffeic acid) in protecting hepatocytes and endothelial cells against triglyceride accumulation and oxidative stress. METHODS: Rat hepatoma and human endothelial cells were exposed to a mixture of oleate/palmitate to mimic the condition of NAFLD and atherosclerosis, respectively. Then, cells were incubated for 24 h in the absence or in the presence of PC or PEOP. Different parameters were evaluated, such as lipid accumulation and oxidative stress-related markers. RESULTS: In hepatic cells, expression of peroxisome proliferator-activated receptors (PPARs) and of stearoyl-CoA desaturase 1 (SCD-1) were assessed as index of lipid metabolism. In endothelial cells, expression of intercellular adhesion molecule-1 (ICAM-1), activation of nuclear factor kappa-B (NF-kB), release of nitric oxide (NO), and wound-healing rate were assessed as index of inflammation. CONCLUSION: PEOP extract ameliorated hepatic lipid accumulation and lipid-dependent oxidative imbalance thus showing potential applications as therapeutic agent tuning down hepatosteatosis and atherosclerosis.


Subject(s)
Endothelial Cells/drug effects , Hepatocytes/drug effects , Oxidative Stress/drug effects , Polyphenols/pharmacology , Animals , Cell Line, Tumor , Cells, Cultured , Endothelial Cells/metabolism , Fatty Acids/adverse effects , Fatty Acids/metabolism , Hepatocytes/metabolism , Humans , Lipid Metabolism , Liver , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Rats
10.
Front Nutr ; 4: 42, 2017.
Article in English | MEDLINE | ID: mdl-28971098

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a major cause of liver-related morbidity and mortality. Oxidative stress and release of pro-inflammatory cytokines, such as tumor necrosis factor α (TNFα), are major consequences of hepatic lipid overload, which can contribute to progression of NAFLD to non-alcoholic steatohepatitis (NASH). Also, mitochondria are involved in the NAFLD pathogenesis for their role in hepatic lipid metabolism. Definitive treatments for NAFLD/NASH are lacking so far. Silybin, the extract of the milk thistle seeds, has previously shown beneficial effects in NAFLD. Sequential exposure of hepatocytes to high concentrations of fatty acids (FAs) and TNFα resulted in fat overload and oxidative stress, which mimic in vitro the progression of NAFLD from simple steatosis (SS) to steatohepatitis (SH). The exposure to 50 µM silybin for 24 h reduced fat accumulation in the model of NAFLD progression. The in vitro progression of NAFLD from SS to SH resulted in reduced hepatocyte viability, increased apoptosis and oxidative stress, reduction in lipid droplet size, and up-regulation of IκB kinase ß-interacting protein and adipose triglyceride lipase expressions. The direct action of silybin on SS or SH cells and the underlying mechanisms were assessed. Beneficial action of silybin was sustained by changes in expression/activity of peroxisome proliferator-activated receptors and enzymes for FA oxidation. Moreover, silybin counteracted the FA-induced mitochondrial damage by acting on complementary pathways: (i) increased the mitochondrial size and improved the mitochondrial cristae organization; (ii) stimulated mitochondrial FA oxidation; (iii) reduced basal and maximal respiration and ATP production in SH cells; (iv) stimulated ATP production in SS cells; and (v) rescued the FA-induced apoptotic signals and oxidative stress in SH cells. We provide new insights about the direct protective effects of the nutraceutic silybin on hepatocytes mimicking in vitro NAFLD progression.

11.
Ann Hepatol ; 16(5): 707-719, 2017.
Article in English | MEDLINE | ID: mdl-28809727

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is the most common liver disorder in industrialized countries and is associated with increased risk of cardiovascular, hepatic and metabolic diseases. Molecular mechanisms on the root of the disrupted lipid homeostasis in NAFLD and potential therapeutic strategies can benefit of in vivo and in vitro experimental models of fatty liver. Here, we describe the high fat diet (HFD)-fed rat in vivo model, and two in vitro models, the primary cultured rat fatty hepatocytes or the FaO rat hepatoma fatty cells, mimicking human NAFLD. Liver steatosis was invariably associated with increased number/size of lipid droplets (LDs) and modulation of expression of genes coding for key genes of lipid metabolism such as peroxisome proliferator-activated receptors (Ppars) and perilipins (Plins). In these models, we tested the anti-steatotic effects of 3,5-L-diiodothyronine (T2), a metabolite of thyroid hormones. T2 markedly reduced triglyceride content and LD size acting on mRNA expression of both Ppars and Plins. T2 also stimulated mitochondrial oxidative metabolism of fatty acids. We conclude that in vivo and especially in vitro models of NAFLD are valuable tools to screen a large number of compounds counteracting the deleterious effect of liver steatosis. Because of the high and negative impact of liver steatosis on human health, ongoing experimental studies from our group are unravelling the ultimate translational value of such cellular models of NAFLD.


Subject(s)
Diiodothyronines/pharmacology , Hepatocytes/drug effects , Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Animals , Cell Line, Tumor , Diet, High-Fat , Disease Models, Animal , Gene Expression Regulation , Hepatocytes/metabolism , Hepatocytes/pathology , High-Throughput Screening Assays , Humans , Lipid Droplets/drug effects , Lipid Droplets/metabolism , Lipid Droplets/pathology , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Liver/metabolism , Liver/pathology , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Rats , Signal Transduction/drug effects , Translational Research, Biomedical/methods
12.
Article in English | MEDLINE | ID: mdl-27626137

ABSTRACT

In vertebrate systems, many endocrine disruptors (EDs) can also interfere with energy and lipid metabolism, thus acting as metabolic disruptors. At the cellular level, these effects are mainly mediated by interactions with nuclear receptors/transcription factors, leading to the modulation of genes involved in lipid homeostasis, as well as by rapid, receptor-independent pathways. Several potential metabolic disruptors are found in aquatic environments. In fish, different EDs have been shown to affect hepatic lipid homeostasis both in vivo and in vitro. However, little information is available in aquatic invertebrates due to our poor knowledge of the regulatory pathways of lipid metabolism. In this work, primary cell cultures from the digestive gland of the bivalve Mytilus galloprovincialis were utilized to investigate the effects of model EDs (bisphenol A (BPA) and perfluorooctane sulphonate (PFOS)) on lipid homeostasis. Both compounds (at 24 and 3h of exposure) increased intracellular lipid and tryglyceride-TAG content, with strongest effects of PFOS at 10-7M. Acyl-CoA oxidase activity was unaffected, whereas some changes in the activity of glycolytic, antioxidant/biotransformation enzymes were observed; however, no clear relationship was found with lipid accumulation. Evaluation of mitochondrial membrane potential Δψm and determination of extracellular TAG content indicate that PFOS interferes with mitochondrial function and lipid secretion, whereas BPA mainly affects lipid secretion. Experiments with specific inhibitors showed that activation of PI-3 kinase and extracellularly regulated mitogen-activated protein kinase (ERK MAPK) plays a key role in mediating lipid accumulation. Mussel digestive gland cells represent a simple in vitro model for screening the metabolic effects of EDs in marine invertebrates.


Subject(s)
Alkanesulfonic Acids/toxicity , Benzhydryl Compounds/toxicity , Digestive System/drug effects , Endocrine Disruptors/toxicity , Energy Metabolism/drug effects , Environmental Monitoring/methods , Fluorocarbons/toxicity , Lipid Metabolism/drug effects , Mytilus/drug effects , Phenols/toxicity , Water Pollutants, Chemical/toxicity , Animals , Cells, Cultured , Digestive System/metabolism , Dose-Response Relationship, Drug , Fish Proteins/antagonists & inhibitors , Fish Proteins/metabolism , Membrane Potential, Mitochondrial , Mitochondria/drug effects , Mitochondria/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Mytilus/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Primary Cell Culture , Protein Kinase Inhibitors/pharmacology , Triglycerides/metabolism
13.
Toxicology ; 361-362: 12-23, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27381660

ABSTRACT

Quartz is a well-known occupational fibrogenic agent able to cause fibrosis and other severe pulmonary diseases such as silicosis and lung cancer. The silicotic pathology owes its severity to the structural and chemo-physical properties of the particles such as shape, size and abundance of surface radicals. In earlier studies, we reported that significant amounts of surface radicals can be generated on crystalline silica by chemical aggression with ascorbic acid (AA), a vitamin naturally abundant in the lung surfactant, and this reaction led to enhanced cytotoxicity and production of inflammatory mediators in a macrophage cell line. However in the lung, other cells acting in the development of silicosis, like fibroblasts and endothelial cells, can come to direct contact with inhaled quartz. We investigated the cytotoxic/pro-inflammatory effects of AA-treated quartz microcrystals (QA) in human primary fibroblasts and endothelial cells as compared to unmodified microcrystals (Q). Our results show that, in fibroblasts, the abundance of surface radicals on quartz microcrystals (Q vs QA) significantly enhanced cell proliferation (with or without co-culture with macrophages), reactive oxygen species (ROS) production, NF-κB nuclear translocation, smooth muscle actin, fibronectin, Bcl-2 and tissue inhibitor of metalloproteinase-1 expression and collagen production. Contrariwise, endothelial cells reacted to the presence of quartz microcrystals independently from the abundance of surface radicals showing similar levels of cytotoxicity, ROS production, cell migration, MCP-1, ICAM-I and fibronectin gene expression when challenged with Q or QA. In conclusion, our in vitro experimental model demonstrates an important and quite unexplored direct contribute of silica surface radicals to fibroblast proliferation and fibrogenic responses.


Subject(s)
Endothelial Cells/drug effects , Fibroblasts/drug effects , Free Radicals/chemistry , Quartz/toxicity , Reactive Oxygen Species/metabolism , Silicon Dioxide/toxicity , Silicosis/pathology , Animals , Cell Line , Cell Proliferation/drug effects , Collagen/biosynthesis , Crystallization , Humans , Mice , NF-kappa B/metabolism , Nitrites/metabolism , Surface Properties , Tissue Inhibitor of Metalloproteinase-1/metabolism , Wound Healing/drug effects
14.
World J Gastroenterol ; 22(26): 6016-26, 2016 Jul 14.
Article in English | MEDLINE | ID: mdl-27468193

ABSTRACT

AIM: To investigate in vitro the therapeutic effect and mechanisms of silybin in a cellular model of hepatic steatosis. METHODS: Rat hepatoma FaO cells were loaded with lipids by exposure to 0.75 mmol/L oleate/palmitate for 3 h to mimic liver steatosis. Then, the steatotic cells were incubated for 24 h with different concentrations (25 to 100 µmol/L) of silybin as phytosome complex with vitamin E. The effects of silybin on lipid accumulation and metabolism, and on indices of oxidative stress were evaluated by absorption and fluorescence microscopy, quantitative real-time PCR, Western blot, spectrophotometric and fluorimetric assays. RESULTS: Lipid-loading resulted in intracellular triglyceride (TG) accumulation inside lipid droplets, whose number and size increased. TG accumulation was mediated by increased levels of peroxisome proliferator-activated receptors (PPARs) and sterol regulatory element-binding protein-1c (SREBP-1c). The lipid imbalance was associated with higher production of reactive oxygen species (ROS) resulting in increased lipid peroxidation, stimulation of catalase activity and activation of nuclear factor kappa-B (NF-κB). Incubation of steatotic cells with silybin 50 µmol/L significantly reduced TG accumulation likely by promoting lipid catabolism and by inhibiting lipogenic pathways, as suggested by the changes in carnitine palmitoyltransferase 1 (CPT-1), PPAR and SREBP-1c levels. The reduction in fat accumulation exerted by silybin in the steatotic cells was associated with the improvement of the oxidative imbalance caused by lipid excess as demonstrated by the reduction in ROS content, lipid peroxidation, catalase activity and NF-κB activation. CONCLUSION: We demonstrated the direct anti-steatotic and anti-oxidant effects of silybin in steatotic cells, thus elucidating at a cellular level the encouraging results demonstrated in clinical and animal studies.


Subject(s)
Antioxidants/pharmacology , Fatty Liver , Hepatocytes/drug effects , Lipid Metabolism/drug effects , Oxidative Stress/drug effects , Silymarin/pharmacology , Vitamin E/pharmacology , Animals , Blotting, Western , Carnitine O-Palmitoyltransferase/drug effects , Carnitine O-Palmitoyltransferase/metabolism , Catalase/drug effects , Catalase/metabolism , Cell Line, Tumor , Cells, Cultured , Fluorometry , Hepatocytes/metabolism , Hepatocytes/pathology , Lipid Droplets/drug effects , Lipid Droplets/metabolism , Lipid Peroxidation/drug effects , Microscopy, Fluorescence , NF-kappa B/drug effects , NF-kappa B/metabolism , Oleic Acid/pharmacology , Palmitates/pharmacology , Peroxisome Proliferator-Activated Receptors/drug effects , Peroxisome Proliferator-Activated Receptors/metabolism , Rats , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Silybin , Spectrophotometry , Sterol Regulatory Element Binding Protein 1/drug effects , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/metabolism
15.
Food Chem Toxicol ; 90: 84-94, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26854922

ABSTRACT

Excess ethanol consumption and fatty acid intake lead to a cumulative effect on liver steatosis through still unclear mechanisms. This study aimed to characterize the lipid homoeostasis alterations under the exposure of hepatocytes to ethanol alone or combined with fatty acids. FaO hepatoma cells were incubated in the absence (C) or in the presence of 100 mM ethanol (EtOH) or 0.35 mM oleate/palmitate (FFA) alone or in the combination (FFA/EtOH). Content of intra- and extra-cellular triglycerides (TAGs) and of lipid droplets (LDs), expression of lipogenic and lipolytic genes, and oxidative stress-related parameters were evaluated. Exposure to either FFAs or EtOH given separately led to steatosis which was augmented when they were combined. Our results show that FFA/EtOH: (i) increased the LD number, but reduced their size compared to separate treatments; (ii) up-regulated PPARγ and SREBP-1c and down-regulated sirtuin-1 (SIRT1); (iii) impaired FFA oxidation; (iv) did not change lipid secretion and oxidative stress. Our findings indicate that one of the major mechanisms of the metabolic interference between ethanol and fat excess is the impairment of FFA oxidation, in addition to lipogenic pathway stimulation. Interestingly, ethanol combined with FFAs led to a shift from macrovesicular to microvesicular steatosis that represents a more dangerous condition.


Subject(s)
Ethanol/pharmacology , Fatty Acids/pharmacology , Lipid Metabolism/drug effects , Animals , Cell Line, Tumor , Fatty Liver , Homeostasis , Rats
16.
Neuromolecular Med ; 18(1): 50-68, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26530396

ABSTRACT

Transgenic mice overexpressing spermine oxidase (SMO) in the cerebral cortex (Dach-SMO mice) showed increased vulnerability to excitotoxic brain injury and kainate-induced epileptic seizures. To investigate the mechanisms by which SMO overexpression leads to increased susceptibility to kainate excitotoxicity and seizure, in the cerebral cortex of Dach-SMO and control mice we assessed markers for astrocyte proliferation and neuron loss, and the ability of kainate to evoke glutamate release from nerve terminals and astrocyte processes. Moreover, we assessed a possible role of astrocytes in an in vitro model of epileptic-like activity in combined cortico-hippocampal slices recorded with a multi-electrode array device. In parallel, as the brain is a major metabolizer of oxygen and yet has relatively feeble protective antioxidant mechanisms, we analyzed the oxidative status of the cerebral cortex of both SMO-overexpressing and control mice by evaluating enzymatic and non-enzymatic scavengers such as metallothioneins. The main findings in the cerebral cortex of Dach-SMO mice as compared to controls are the following: astrocyte activation and neuron loss; increased oxidative stress and activation of defense mechanisms involving both neurons and astrocytes; increased susceptibility to kainate-evoked cortical epileptogenic activity, dependent on astrocyte function; appearance of a glutamate-releasing response to kainate from astrocyte processes due to activation of Ca(2+)-permeable AMPA receptors in Dach-SMO mice. We conclude that reactive astrocytosis and activation of glutamate release from astrocyte processes might contribute, together with increased reactive oxygen species production, to the vulnerability to kainate excitotoxicity in Dach-SMO mice. This mouse model with a deregulated polyamine metabolism would shed light on roles for astrocytes in increasing vulnerability to excitotoxic neuron injury.


Subject(s)
Astrocytes/drug effects , Kainic Acid/pharmacology , Nerve Tissue Proteins/physiology , Neurotoxins/pharmacology , Oxidoreductases Acting on CH-NH Group Donors/physiology , Seizures/chemically induced , Animals , Aspartic Acid/metabolism , Astrocytes/pathology , Benzodiazepines/pharmacology , Biogenic Polyamines/metabolism , Calcium/metabolism , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Enzyme Induction , Genetic Predisposition to Disease , Gliosis/genetics , Hippocampus/enzymology , Hippocampus/pathology , Hippocampus/physiopathology , Male , Metallothionein/physiology , Mice , Mice, Neurologic Mutants , Mice, Transgenic , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/pathology , Oxidative Stress , Oxidoreductases Acting on CH-NH Group Donors/biosynthesis , Oxidoreductases Acting on CH-NH Group Donors/genetics , Receptors, AMPA/drug effects , Receptors, AMPA/physiology , Recombinant Fusion Proteins/biosynthesis , Seizures/genetics , Seizures/physiopathology , Synaptosomes/drug effects , Synaptosomes/physiology , Up-Regulation , Polyamine Oxidase
17.
World J Biol Psychiatry ; 17(3): 198-209, 2016 04.
Article in English | MEDLINE | ID: mdl-26469135

ABSTRACT

OBJECTIVES: Oxidative stress seems to be involved in Rett syndrome (RTT). The aim of this study was to assess the antioxidant status in RTT children with MECP2 gene mutations with respect to healthy controls, and to explore novel blood antioxidant markers for RTT severity. METHODS: In erythrocytes from RTT females aged 2-14 years (n = 27) and age-matched controls (n = 27), we measured the levels of malonaldehyde and the activity of two antioxidant enzymes, Cu/Zn-superoxide dismutase and catalase, by spectrophotometric assays. In leukocytes, the expression of metallothioneins, the main non-enzymatic antioxidants, was assessed by real-time RT-PCR. In nine selected RTT children, methylome analysis was also performed. RESULTS: Blood of RTT patients showed increased lipid peroxidation and a dysregulated pattern of MT expression, while enzymatic activities did not change significantly with respect to controls. Moreover, we observed no epigenetic dysregulation in CpG-enriched promoter regions of the analysed genes but significant hypomethylation in the random loci. CONCLUSIONS: As the haematic level of MT-1A directly correlates with the phenotype severity, this metallothionein can represent a marker for RTT severity. Moreover, the attempt to link the level of blood oxidative stress with MECP2 mutation and specific clinical features led us to draw some interesting conclusions.


Subject(s)
DNA Methylation , Metallothionein/metabolism , Methyl-CpG-Binding Protein 2/genetics , Oxidative Stress , Rett Syndrome/genetics , Adolescent , Biomarkers , Case-Control Studies , Child , Child, Preschool , DNA Mutational Analysis , Female , Humans , Metallothionein/genetics , Mutation , Regression Analysis
18.
Stem Cell Res Ther ; 6: 245, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26651832

ABSTRACT

INTRODUCTION: Systemic delivery of bone marrow-derived mesenchymal stem cells (MSC) seems to be of benefit in the treatment of multiple sclerosis (MS), an autoimmune disease of the central nervous system (CNS) sustained by migration of T cells across the brain blood barrier (BBB) and subsequent induction of inflammatory lesions into CNS. MSC have been found to modulate several effector functions of T cells. In this study, we investigated the effects of MSC on adhesion molecules and receptors on T cell surface that sustain their transendothelial migration. METHODS: We used different co-culture methods combined with real-time PCR and flow cytometry to evaluate the expression both at the mRNA and at the plasma-membrane level of α4 integrin, ß2 integrin, ICAM-1 and CXCR3. In parallel, we assessed if MSC are able to modulate expression of adhesion molecules on the endothelial cells that interact with T cells during their transendothelial migration. RESULTS: Our in vitro analyses revealed that MSC: (i) inhibit proliferation and activation of both peripheral blood mononuclear cells (PBMC) and CD3(+)-selected lymphocytes through the release of soluble factors; (ii) exert suppressive effects on those surface molecules highly expressed by activated lymphocytes and involved in transendothelial migration; (iii) inhibit CXCL10-driven chemotaxis of CD3(+) cells; (iv) down-regulated expression of adhesion molecules on endothelial cells. CONCLUSIONS: Taken together, these data demonstrate that the immunosuppressive effect of MSC does not exclusively depends on their anti-proliferative activity on T cells, but also on the impairment of leukocyte migratory potential through the inhibition of the adhesion molecules and receptors that are responsible for T cell trafficking across BBB. This could suggest a new mechanism through which MSC modulate T cell responses.


Subject(s)
Cell Adhesion Molecules/metabolism , Mesenchymal Stem Cells/immunology , T-Lymphocytes/immunology , Cell Adhesion Molecules/genetics , Cell Movement/immunology , Cell Proliferation , Chemokine CXCL10/metabolism , Chemotaxis, Leukocyte , Coculture Techniques , Endothelial Cells/cytology , Endothelial Cells/immunology , Gene Expression , Humans , Immunosuppression Therapy , Lymphocyte Activation , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism , T-Lymphocytes/cytology
19.
Front Physiol ; 6: 418, 2015.
Article in English | MEDLINE | ID: mdl-26793120

ABSTRACT

Adipose tissue, dietary lipids and de novo lipogenesis are sources of hepatic free fatty acids (FFAs) that are stored in lipid droplets (LDs) as triacylglycerols (TAGs). Destiny of TAGs stored in LDs is determined by LD proteomic equipment. When adipose triglyceride lipase (ATGL) localizes at LD surface the lipid mobilization is stimulated. In this work, an in vitro model of cultured rat hepatocytes mimicking a mild steatosis condition was used to investigate the direct lipid-lowering action of iodothyronines, by focusing, in particular, on LD-associated proteins, FFA oxidation and lipid secretion. Our results demonstrate that in "steatotic" hepatocytes iodothyronines reduced the lipid excess through the recruitment of ATGL on LD surface, and the modulation of the LD-associated proteins Rab18 and TIP47. As an effect of ATGL recruitment, iodothyronines stimulated the lipid mobilization from LDs then followed by the up-regulation of carnitine-palmitoyl-transferase (CPT1) expression and the stimulation of cytochrome-c oxidase (COX) activity that seems to indicate a stimulation of mitochondrial function. The lipid lowering action of iodothyronines did not depend on increased TAG secretion. On the basis of our data, ATGL could be indicated as an early mediator of the lipid-lowering action of iodothyronines able to channel hydrolyzed FFAs toward mitochondrial beta-oxidation rather than secretion.

20.
Drug Alcohol Depend ; 143: 112-9, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25107314

ABSTRACT

BACKGROUND: Oxidative stress is implicated in pathogenesis of alcoholic liver disease (ALD). This study investigated the possible correlation among the erythrocyte indices of oxidative stress, the leukocyte panels of antioxidant proteins (metallothioneins), the serum biochemical parameters and the liver steatosis grade. METHODS: A total of 118 cases including 60 alcoholic subjects and 58 controls were enrolled. All the alcoholic subjects were screened for body mass index (BMI), liver steatosis, and blood chemistry and serology. The level of oxidative stress and oxidative stress-related parameters were measured in the blood and correlated with clinical findings. RESULTS: Alcoholic subjects showed higher BMI, moderate/severe hepatic steatosis, increase in the levels of triglycerides, cholesterol, glucose, γ-glutamyl-transpeptidase (GGT), alanine aminotransferase (ALT), bilirubin, alpha 1 and beta 2 globulins, iron and a decrease in the levels of aspartate aminotransferase (AST) and beta 1 globulin with respect to the reference values. Moreover, alcoholic subjects showed: (i) an increase in Thiobarbituric Acid Reactive Substance (TBARS) content representing a good estimation of global oxidative stress; (ii) a stimulation of the activities of the antioxidant enzymes catalase and SOD; (iii) a modulation of expression of metallothioneins, with a down-regulation of MT-1A and an up-regulation of MT-1E isoforms. CONCLUSIONS: Our data suggest that alcoholism is strongly associated with altered pattern of blood metallothioneins; this parameter combined with the score calculated by an ad hoc implemented algorithm (HePaTest) could offer a non-invasive alternative approach for evaluating alcohol-related damages and could be used in follow-up of alcoholic patients.


Subject(s)
Alcoholism/blood , Antioxidants/metabolism , Liver Diseases, Alcoholic/blood , Oxidative Stress , Adolescent , Adult , Alcoholism/complications , Case-Control Studies , Erythrocyte Indices , Fatty Liver/blood , Fatty Liver/complications , Humans , Leukocytes/metabolism , Lipid Peroxidation , Liver Diseases, Alcoholic/complications , Liver Diseases, Alcoholic/diagnostic imaging , Male , Metallothionein/biosynthesis , Middle Aged , Ultrasonography , Up-Regulation , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...