Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Nat Cancer ; 5(1): 16-29, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38273023

ABSTRACT

Metastasis formation is a complex process, involving multiple crucial steps, which are controlled by different regulatory mechanisms. In this context, the contribution of cancer metabolism to the metastatic cascade is being increasingly recognized. This Review focuses on changes in lipid metabolism that contribute to metastasis formation in solid tumors. We discuss the molecular mechanisms by which lipids induce a pro-metastatic phenotype and explore the role of lipids in response to oxidative stress and as signaling molecules. Finally, we reflect on potential avenues to target lipid metabolism to improve the treatment of metastatic cancers.


Subject(s)
Neoplasms , Humans , Signal Transduction , Oxidative Stress , Lipids
2.
Free Radic Biol Med ; 208: 285-298, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37619957

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by progressive loss of motor neurons, systemic hypermetabolism, and inflammation. In this context, oxylipins have been investigated as signaling molecules linked to neurodegeneration, although their specific role in ALS remains unclear. Importantly, most methods focused on oxylipin analysis are based on low-resolution mass spectrometry, which usually confers high sensitivity, but not great accuracy for molecular characterization, as provided by high-resolution MS (HRMS). Here, we established an ultra-high performance liquid chromatography HRMS (LC-HRMS) method for simultaneous analysis of 126 oxylipins in plasma. Intra- and inter-day method validation showed high sensitivity (0.3-25 pg), accuracy and precision for more than 90% of quality controls. This method was applied in plasma of ALS rats overexpressing the mutant human Cu/Zn-superoxide dismutase gene (SOD1-G93A) at asymptomatic (ALS 70 days old) and symptomatic stages (ALS 120 days old), and their respective age-matched wild type controls. From the 56 oxylipins identified in plasma, 17 species were significantly altered. Remarkably, most of oxylipins linked to inflammation and oxidative stress derived from arachidonic acid (AA), like prostaglandins and mono-hydroxides, were increased in ALS 120 d rats. In addition, ketones derived from AA and linoleic acid (LA) were increased in both WT 120 d and ALS 120 d groups, supporting that age also modulates oxylipin metabolism in plasma. Interestingly, the LA-derived diols involved in fatty acid uptake and ß-oxidation, 9(10)-DiHOME and 12(13)-DiHOME, were decreased in ALS 120 d rats and showed significant synergic effects between age and disease factors. In summary, we validated a high-throughput LC-HRMS method for oxylipin analysis and provided a comprehensive overview of plasma oxylipins involved in ALS disease progression. Noteworthy, the oxylipins altered in plasma have potential to be investigated as biomarkers for inflammation and hypermetabolism in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Rats , Humans , Animals , Mice , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Oxylipins , Mass Spectrometry , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Inflammation , Disease Models, Animal , Mice, Transgenic , Superoxide Dismutase/genetics
3.
Cancer Cell ; 41(7): 1204-1206, 2023 07 10.
Article in English | MEDLINE | ID: mdl-37244258

ABSTRACT

Cholesterol is essential for the ability of cytotoxic T cells to eliminate cancer cells. In this issue of Cancer Cell, Yan et al. reveal how intra-tumoral cholesterol deficiency contributes to T cell exhaustion by inhibiting mTORC1 signaling. Moreover, they demonstrate that increasing cholesterol levels in chimeric antigen receptor (CAR)-T cells by blocking liver X receptor (LXR) leads to improved anti-tumor function.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , Tumor Microenvironment , Immunotherapy, Adoptive , Neoplasms/therapy , T-Lymphocytes, Cytotoxic
4.
Article in English | MEDLINE | ID: mdl-36535597

ABSTRACT

Total absence of adipose tissue (lipoatrophy) is associated with the development of severe metabolic disorders including hepatomegaly and fatty liver. Here, we sought to investigate the impact of severe lipoatrophy induced by deletion of peroxisome proliferator-activated receptor gamma (PPARγ) exclusively in adipocytes on lipid metabolism in mice. Untargeted lipidomics of plasma, gastrocnemius and liver uncovered a systemic depletion of the essential linoleic (LA) and α-linolenic (ALA) fatty acids from several lipid classes (storage lipids, glycerophospholipids, free fatty acids) in lipoatrophic mice. Our data revealed that such essential fatty acid depletion was linked to increased: 1) capacity for liver mitochondrial fatty acid ß-oxidation (FAO), 2) citrate synthase activity and coenzyme Q content in the liver, 3) whole-body oxygen consumption and reduced respiratory exchange rate in the dark period, and 4) de novo lipogenesis and carbon flux in the TCA cycle. The key role of de novo lipogenesis in hepatic steatosis was evidenced by an accumulation of stearic, oleic, sapienic and mead acids in liver. Our results thus indicate that the simultaneous activation of the antagonic processes FAO and de novo lipogenesis in liver may create a futile metabolic cycle leading to a preferential depletion of LA and ALA. Noteworthy, this previously unrecognized cycle may also explain the increased energy expenditure displayed by lipoatrophic mice, adding a new piece to the metabolic regulation puzzle in lipoatrophies.


Subject(s)
Fatty Liver , Lipogenesis , Animals , Mice , Substrate Cycling , Lipid Metabolism , Fatty Liver/metabolism , alpha-Linolenic Acid/metabolism
5.
Biochim Biophys Acta Mol Basis Dis ; 1868(6): 166371, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35218894

ABSTRACT

Cardiovascular manifestations account for marked morbi-mortality in autosomal dominant polycystic kidney disease (ADPKD). Pkd1- and Pkd2-deficient mice develop cardiac dysfunction, however the underlying mechanisms remain largely unclear. It is unknown whether impairment of polycystin-1 cleavage at the G-protein-coupled receptor proteolysis site, a significant ADPKD mutational mechanism, is involved in this process. We analyzed the impact of polycystin-1 cleavage on heart metabolism using Pkd1V/V mice, a model unable to cleave this protein and with early cardiac dysfunction. Pkd1V/V hearts showed lower levels of glucose and amino acids and higher lipid levels than wild-types, as well as downregulation of p-AMPK, p-ACCß, CPT1B-Cpt1b, Ppara, Nppa and Acta1. These findings suggested decreased fatty acid ß-oxidation, which was confirmed by lower oxygen consumption by Pkd1V/V isolated mitochondria using palmitoyl-CoA. Pkd1V/V hearts also presented increased oxygen consumption in response to glucose, suggesting that alternative substrates may be used to generate energy. Pkd1V/V hearts displayed a higher density of decreased-size mitochondria, a finding associated with lower MFN1, Parkin and BNIP3 expression. These derangements were correlated with increased apoptosis and inflammation but not hypertrophy. Notably, Pkd1V/V neonate cardiomyocytes also displayed shifts in oxygen consumption and p-AMPK downregulation, suggesting that, at least partially, the metabolic alterations are not induced by kidney dysfunction. Our findings reveal that disruption of polycystin-1 cleavage leads to cardiac metabolic rewiring in mice, expanding the understanding of heart dysfunction associated with Pkd1 deficiency and likely with human ADPKD.


Subject(s)
Polycystic Kidney, Autosomal Dominant , TRPP Cation Channels , Animals , Heart , Mice , Mitochondria/metabolism , Mutation , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism
7.
Nat Prod Res ; 36(10): 2565-2569, 2022 May.
Article in English | MEDLINE | ID: mdl-33749461

ABSTRACT

Hancornia speciosa Gomes (HS) is a Brazilian fruit tree used in inflammatory disorders by folk medicine. Here we sought to investigate the antioxidant and hepatoprotective properties of HS fruit juice in acetaminophen-induced hepatoxicity model in rats, as well as to perform a comprehensive chemical characterization of phenolics and metals by mass spectrometry. HS showed hepatoprotective and antioxidant activity by decreasing MDA and liver injury markers at healthy control levels. In addition, liver histopathological analysis revealed that HS decreases hepatocellular degeneration. Chemical characterization revealed 16 different phenolics, being chlorogenic acid (150 ± 5 µg/g) and rutin (120 ± 8 µg/g) the major phenolics in HS. Among the 14 micronutrients we identified, zinc and boron were the most abundant metals detected in HS. In line with previous studies involving liver diseases, our data supports evidence that such phenolics and metals present in HS may prevent liver injury induced by acetaminophen.


Subject(s)
Apocynaceae , Chemical and Drug Induced Liver Injury , Acetaminophen/toxicity , Animals , Antioxidants/pharmacology , Apocynaceae/chemistry , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/prevention & control , Fruit and Vegetable Juices , Liver , Phenols/pharmacology , Plant Extracts/chemistry , Plant Extracts/pharmacology , Rats
8.
Nanomedicine ; 36: 102418, 2021 08.
Article in English | MEDLINE | ID: mdl-34171470

ABSTRACT

Radiation induces the generation of platelet-activating factor receptor (PAF-R) ligands, including PAF and oxidized phospholipids. Alternatively, PAF is also synthesized by the biosynthetic enzymes lysophosphatidylcholine acyltransferases (LPCATs) which are expressed by tumor cells including melanoma. The activation of PAF-R by PAF and oxidized lipids triggers a survival response protecting tumor cells from radiation-induced cell death, suggesting the involvement of the PAF/PAF-R axis in radioresistance. Here, we investigated the role of LPCATs in the melanoma cell radiotherapy response. LPCAT is a family of four enzymes, LPCAT1-4, and modular nucleic acid nanoparticles (NANPs) allowed for the simultaneous silencing of all four LPCATs. We found that the in vitro simultaneous silencing of all four LPCAT transcripts by NANPs enhanced the therapeutic effects of radiation in melanoma cells by increasing cell death, reducing long-term cell survival, and activating apoptosis. Thus, we propose that NANPs are an effective strategy for improving radiotherapy efficacy in melanomas.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase , Gene Silencing , Melanoma , Nanoparticles , Neoplasm Proteins , Nucleic Acids , 1-Acylglycerophosphocholine O-Acyltransferase/antagonists & inhibitors , 1-Acylglycerophosphocholine O-Acyltransferase/biosynthesis , Cell Line, Tumor , Humans , Melanoma/drug therapy , Melanoma/enzymology , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Nucleic Acids/chemistry , Nucleic Acids/pharmacology
9.
Article in English | MEDLINE | ID: mdl-34004356

ABSTRACT

The nutrient sensors peroxisome proliferator-activated receptor γ (PPARγ) and mechanistic target of rapamycin complex 1 (mTORC1) closely interact in the regulation of adipocyte lipid storage. The precise mechanisms underlying this interaction and whether this extends to other metabolic processes and the endocrine function of adipocytes are still unknown. We investigated herein the involvement of mTORC1 as a mediator of the actions of the PPARγ ligand rosiglitazone in subcutaneous inguinal white adipose tissue (iWAT) mass, endocrine function, lipidome, transcriptome and branched-chain amino acid (BCAA) metabolism. Mice bearing regulatory associated protein of mTOR (Raptor) deletion and therefore mTORC1 deficiency exclusively in adipocytes and littermate controls were fed a high-fat diet supplemented or not with the PPARγ agonist rosiglitazone (30 mg/kg/day) for 8 weeks and evaluated for iWAT mass, lipidome, transcriptome (Rnaseq), respiration and BCAA metabolism. Adipocyte mTORC1 deficiency not only impaired iWAT adiponectin transcription, synthesis and secretion, PEPCK mRNA levels, triacylglycerol synthesis and BCAA oxidation and mRNA levels of related proteins but also completely blocked the upregulation in these processes induced by pharmacological PPARγ activation with rosiglitazone. Mechanistically, adipocyte mTORC1 deficiency impairs PPARγ transcriptional activity by reducing PPARγ protein content, as well as by downregulating C/EBPα, a co-partner and facilitator of PPARγ. In conclusion, mTORC1 and PPARγ are essential partners involved in the regulation of subcutaneous adipose tissue adiponectin production and secretion and BCAA oxidative metabolism.


Subject(s)
Adiponectin/metabolism , Amino Acids, Branched-Chain/metabolism , Glycerol/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , PPAR gamma/metabolism , Subcutaneous Fat/metabolism , Up-Regulation , Animals , Mice , Oxidation-Reduction
10.
J Nutr Biochem ; 87: 108519, 2021 01.
Article in English | MEDLINE | ID: mdl-33017610

ABSTRACT

Dietary sugar is an important determinant of the development and progression of nonalcoholic fatty liver disease (NAFLD). However, the molecular mechanisms underlying the deleterious effects of sugar intake on NAFLD under energy-balanced conditions are still poorly understood. Here, we provide a comprehensive analysis of the liver lipidome and mechanistic insights into the pathogenesis of NAFLD induced by the chronic consumption of high-sugar diet (HSD). Newly weaned male Wistar rats were fed either a standard chow diet or an isocaloric HSD for 18 weeks. Livers were harvested for histological, oxidative stress, gene expression, and lipidomic analyses. Intake of HSD increased oxidative stress and induced severe liver injury, microvesicular steatosis, and ballooning degeneration of hepatocytes. Using untargeted lipidomics, we identified and quantified 362 lipid species in the liver. Rats fed with HSD displayed increased hepatic levels of triacylglycerol enriched in saturated and monounsaturated fatty acids, lipids related to mitochondrial function/structure (phosphatidylglycerol, cardiolipin, and ubiquinone), and acylcarnitine (an intermediate lipid of fatty acid beta-oxidation). HSD-fed animals also presented increased levels of some species of membrane lipids and a decreased content of phospholipids containing omega-6 fatty acids. These changes in the lipidome were associated with the downregulation of genes involved in fatty acid oxidation in the liver. In conclusion, our data suggest that the chronic intake of a HSD, even under isocaloric conditions, induces lipid overload, and inefficient/impaired fatty acid oxidation in the liver. Such events lead to marked disturbance in hepatic lipid metabolism and the development of NAFLD.


Subject(s)
Diet, Carbohydrate Loading/adverse effects , Lipid Metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Lipidomics , Male , Metabolic Networks and Pathways , Rats, Wistar
11.
Langmuir ; 36(48): 14514-14529, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33210931

ABSTRACT

Two commercial exogenous pulmonary surfactants, Curosurf and Survanta, are investigated. Their thermotropic behavior and associated structural changes for the samples in bulk are characterized and described. For Survanta, the obtained results of differential scanning calorimetry showed a thermogram with three peaks on heating and only a single peak on cooling. Curosurf on the other hand, presents calorimetric thermograms with only one peak in both the heating and cooling scans. This distinct thermotropic behavior between the two pulmonary surfactants, a consequence of their particular compositions, is associated with structural changes that were evaluated by simultaneous small- and wide-angle X-ray scattering experiments with in situ temperature variation. Interestingly, for temperatures below ∼35 °C for Curosurf and ∼53 °C for Survanta, the scattering data indicated the coexistence of two lamellar phases with different carbon chain organizations. For temperatures above these limits, the coexistence of phases disappears, giving rise to a fluid phase in both pulmonary surfactants, with multilamelar vesicles for Curosurf and unilamellar vesicles for Survanta. This process is quasi-reversible under cooling, and advanced data analysis for the scattering data indicated differences in the structural and elastic properties of the pulmonary surfactants. The detailed and systematic investigation shown in this work expands on the knowledge of the structure and thermodynamic behavior of Curosurf and Survanta, being relevant from both physiological and biophysical perspectives and also providing a basis for further studies on other types of pulmonary surfactants.


Subject(s)
Pulmonary Surfactants , Animals , Calorimetry, Differential Scanning , Cattle , Lung , Surface-Active Agents , Swine , Thermodynamics
12.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1865(10): 158776, 2020 10.
Article in English | MEDLINE | ID: mdl-32738301

ABSTRACT

Palmitoleic acid (POA, 16:1n-7) is a lipokine that has potential nutraceutical use to treat non-alcoholic fatty liver disease. We tested the effects of POA supplementation (daily oral gavage, 300 mg/Kg, 15 days) on murine liver inflammation induced by a high fat diet (HFD, 59% fat, 12 weeks). In HFD-fed mice, POA supplementation reduced serum insulin and improved insulin tolerance compared with oleic acid (OA, 300 mg/Kg). The livers of POA-treated mice exhibited less steatosis and inflammation than those of OA-treated mice with lower inflammatory cytokine levels and reduced toll-like receptor 4 protein content. The anti-inflammatory effects of POA in the liver were accompanied by a reduction in liver macrophages (LM, CD11c+; F4/80+; CD86+), an effect that could be triggered by peroxisome proliferator activated receptor (PPAR)-γ, a lipogenic transcription factor upregulated in livers of POA-treated mice. We also used HFD-fed mice with selective deletion of PPAR-γ in myeloid cells (PPAR-γ KOLyzCre+) to test whether the beneficial anti-inflammatory effects of POA are dependent on macrophages PPAR-γ. POA-mediated improvement of insulin tolerance was tightly dependent on myeloid PPAR-γ, while POA anti-inflammatory actions including the reduction in liver inflammatory cytokines were preserved in mice bearing myeloid cells deficient in PPAR-γ. This overlapped with increased CD206+ (M2a) cells and downregulation of CD86+ and CD11c+ liver macrophages. Moreover, POA supplementation increased hepatic AMPK activity and decreased expression of the fatty acid binding scavenger receptor, CD36. We conclude that POA controls liver inflammation triggered by fat accumulation through induction of M2a macrophages independently of myeloid cell PPAR-γ.


Subject(s)
Fatty Acids, Monounsaturated/pharmacology , Inflammation/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , PPAR gamma/genetics , AMP-Activated Protein Kinase Kinases , Animals , B7-2 Antigen/genetics , CD11c Antigen/genetics , Diet, High-Fat/adverse effects , Fatty Acids, Monounsaturated/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Insulin Resistance/genetics , Lectins, C-Type/genetics , Liver/drug effects , Liver/metabolism , Liver/pathology , Mannose Receptor , Mannose-Binding Lectins/genetics , Mice , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Oleic Acid/metabolism , Oleic Acid/pharmacology , Protein Kinases/genetics , Receptors, Cell Surface/genetics
13.
An Acad Bras Cienc ; 92(1): e20190491, 2020.
Article in English | MEDLINE | ID: mdl-32401840

ABSTRACT

The Commelina erecta L. (C. erecta) also known as erva-de-santa-luzia is reported by local population to have medical properties against some pathological conditions. In this study, two extracts of C. erecta leaves (aqueous and ethanolic) were phytochemically analysed and evaluated for their in-vitro antioxidant activities by DPPH, TBARS, NO assays and cell viability assays. The ultra-high performance liquid chromatography followed by tandem mass spectrometry analysis showed the presence of rutin and caffeic acid in aqueous and ethanolic extract. The total polyphenols in aqueous and ethanolic extracts found were 142.7 ± 3.0 and 123.1 ± 5.8 µg/mL of GAE, respectively. The ethanolic extract (5 mg/mL) inhibits TBARS by 33.8%, and the aqueous extract (5 mg/mL) exhibited scavenger property against nitric oxide derivatives to an extent of 77.8%. In cell culture, both extracts improved cell survivability under H2O2 induced oxidative stress. Thus, C. erecta extract is a good candidate to become a phytotherapic medicine.


Subject(s)
Antioxidants/pharmacology , Caffeic Acids/analysis , Chromatography, High Pressure Liquid/methods , Commelina/chemistry , Plant Extracts/pharmacology , Rutin/analysis , Animals , Cell Culture Techniques , Humans , Hydrogen Peroxide/pharmacokinetics , Oxidative Stress/drug effects , Phenols/analysis , Phytochemicals/analysis , Plant Leaves/chemistry , Polyphenols/analysis , Tandem Mass Spectrometry/methods
14.
Mol Nutr Food Res ; 64(7): e1900833, 2020 04.
Article in English | MEDLINE | ID: mdl-31978277

ABSTRACT

SCOPE: Glucose homeostasis and progression of nonalcoholic fatty liver disease (NAFLD) and hepatomegaly in severe lipoatrophic mice and their modulation by intake of a diet rich in omega 3 (n-3) fatty acids (HFO) are evaluated. METHODS AND RESULTS: Severe lipoatrophic mice induced by PPAR-γ deletion exclusively in adipocytes (A-PPARγ KO) and littermate controls (A-PPARγ WT) are evaluated for glucose homeostasis and liver mass, proteomics, lipidomics, inflammation, and fibrosis. Lipoatrophic mice are heavier than controls, severely glucose intolerant, and hyperinsulinemic, and develop NAFLD characterized by increased liver glycogen, triacylglycerol, and diacylglycerol contents, mitotic index, apoptosis, inflammation, steatosis score, fibrosis, and fatty acid synthase (FAS) content and activity. Lipoatrophic mice also display liver enrichment with monounsaturated in detriment of polyunsaturated fatty acids including n-3 fatty acids, and increased content of cardiolipin, a tetracyl phospholipid exclusively found at the mitochondria inner membrane. Administration of a high-fat diet rich in n-3 fatty acids (HFO) to lipoatrophic mice enriches liver with n-3 fatty acids, reduces hepatic steatosis, FAS content and activity, apoptosis, inflammation, and improves glucose homeostasis. CONCLUSION: Diet enrichment with n-3 fatty acids improves glucose homeostasis and reduces liver steatosis and inflammation without affecting hepatomegaly in severe lipoatrophic mice.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Insulin Resistance , Lipodystrophy/complications , Non-alcoholic Fatty Liver Disease/diet therapy , Adipocytes/metabolism , Animals , Diet, High-Fat , Glucose/metabolism , Homeostasis/drug effects , Male , Mice, Knockout , Mice, Transgenic , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , PPAR gamma/genetics
15.
Mol Nutr Food Res, 1900833, jan. 2020
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: bud-2936

ABSTRACT

Scope Glucose homeostasis and progression of nonalcoholic fatty liver disease (NAFLD) and hepatomegaly in severe lipoatrophic mice and their modulation by intake of a diet rich in omega 3 (n-3) fatty acids (HFO) are evaluated. Methods and results Severe lipoatrophic mice induced by PPAR-gama deletion exclusively in adipocytes (A-PPARgama KO) and littermate controls (A-PPARgama WT) are evaluated for glucose homeostasis and liver mass, proteomics, lipidomics, inflammation, and fibrosis. Lipoatrophic mice are heavier than controls, severely glucose intolerant, and hyperinsulinemic, and develop NAFLD characterized by increased liver glycogen, triacylglycerol, and diacylglycerol contents, mitotic index, apoptosis, inflammation, steatosis score, fibrosis, and fatty acid synthase (FAS) content and activity. Lipoatrophic mice also display liver enrichment with monounsaturated in detriment of polyunsaturated fatty acids including n-3 fatty acids, and increased content of cardiolipin, a tetracyl phospholipid exclusively found at the mitochondria inner membrane. Administration of a high-fat diet rich in n-3 fatty acids (HFO) to lipoatrophic mice enriches liver with n-3 fatty acids, reduces hepatic steatosis, FAS content and activity, apoptosis, inflammation, and improves glucose homeostasis. Conclusion Diet enrichment with n-3 fatty acids improves glucose homeostasis and reduces liver steatosis and inflammation without affecting hepatomegaly in severe lipoatrophic mice.

16.
Mol. Nutr. Food. Res. ; : 1900833, 2020.
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: but-ib17431

ABSTRACT

Scope Glucose homeostasis and progression of nonalcoholic fatty liver disease (NAFLD) and hepatomegaly in severe lipoatrophic mice and their modulation by intake of a diet rich in omega 3 (n-3) fatty acids (HFO) are evaluated. Methods and results Severe lipoatrophic mice induced by PPAR-gama deletion exclusively in adipocytes (A-PPARgama KO) and littermate controls (A-PPARgama WT) are evaluated for glucose homeostasis and liver mass, proteomics, lipidomics, inflammation, and fibrosis. Lipoatrophic mice are heavier than controls, severely glucose intolerant, and hyperinsulinemic, and develop NAFLD characterized by increased liver glycogen, triacylglycerol, and diacylglycerol contents, mitotic index, apoptosis, inflammation, steatosis score, fibrosis, and fatty acid synthase (FAS) content and activity. Lipoatrophic mice also display liver enrichment with monounsaturated in detriment of polyunsaturated fatty acids including n-3 fatty acids, and increased content of cardiolipin, a tetracyl phospholipid exclusively found at the mitochondria inner membrane. Administration of a high-fat diet rich in n-3 fatty acids (HFO) to lipoatrophic mice enriches liver with n-3 fatty acids, reduces hepatic steatosis, FAS content and activity, apoptosis, inflammation, and improves glucose homeostasis. Conclusion Diet enrichment with n-3 fatty acids improves glucose homeostasis and reduces liver steatosis and inflammation without affecting hepatomegaly in severe lipoatrophic mice.

17.
Chem Res Toxicol ; 32(10): 2028-2041, 2019 10 21.
Article in English | MEDLINE | ID: mdl-31496224

ABSTRACT

Radical mediated oxidation of polyunsaturated fatty acids (PUFA) is known to generate a series of polyoxygenated cyclic products (PUFA-On, n ≥ 3). Here, we describe the characterization of glutathione (GSH) conjugates bound to polyoxygenated docosahexaenoic (DHA-On, n = 3-9), arachidonic (ARA-On, n = 3-7), α-linolenic (ALA-O3), and linoleic (LA-O3) acid species. Similar conjugates were also characterized for N-acetylcysteine (NAC) and Cu,Zn-superoxide dismutase (SOD1). Extensive LC-MS/MS characterization using a synthetic α-linolenic hydroxy-endoperoxide (ALA-O3) derivative revealed at least two types of mechanisms leading to thiol adduction: a mechanism involving the nucleophilic attack by thiolate anion on 1,2-dioxolane to form a sulfenate ester-bonded conjugate and a mechanism involving cleavage of the dioxolane to form a α,ß-unsaturated carbonyl followed by the Michael addition reaction. Finally, we detected a GSH conjugate with hydroxy-endoperoxide derived from linoleic acid (LA-O3) in mice liver. In summary, our study reveals the formation of a series of thiol conjugates that are bound to highly oxygenated PUFA species. GSH conjugates described in our study may potentially play relevant roles in redox and inflammatory processes, especially under high oxygen tension conditions.


Subject(s)
Fatty Acids, Unsaturated/chemistry , Glutathione/chemistry , Animals , Chromatography, Liquid , Fatty Acids, Unsaturated/metabolism , Glutathione/isolation & purification , Glutathione/metabolism , Liver/chemistry , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Oxidation-Reduction , Peroxides/chemistry , Peroxides/metabolism , Tandem Mass Spectrometry
18.
Biosci Rep ; 39(4)2019 04 30.
Article in English | MEDLINE | ID: mdl-30867255

ABSTRACT

Microglial activation by oleate and palmitate differentially modulates brain inflammatory status. However, the metabolic reprogramming supporting these reactive phenotypes remains unknown. Employing real-time metabolic measurements and lipidomic analysis, we show that both fatty acids promote microglial oxidative metabolism, while lipopolysaccharide (LPS) enhances glycolytic rates. Interestingly, oleate treatment was followed by enrichment in storage lipids bound to polyunsaturated fatty acids (PUFA), in parallel with protection against oxidative imbalance. Palmitate, in turn, induced a distinct lipid distribution defined by PUFA linked to membrane phospholipids, which are more susceptible to lipid peroxidation and inflammatory signaling cascades. This distribution was mirrored by LPS treatment, which led to a strong pro-inflammatory phenotype in microglia. Thus, although both oleate and palmitate preserve mitochondrial function, a contrasting lipid distribution supports differences in fatty acid-induced neuroinflammation. These data reinforce the concept that reactive microglial profiles are achieved by stimulus-evoked remodeling in cell metabolism.


Subject(s)
Brain/pathology , Microglia/metabolism , Mitochondria/metabolism , Oleic Acid/metabolism , Palmitates/metabolism , Animals , Cell Line , Glycolysis/physiology , Inflammation/pathology , Lipid Peroxidation/physiology , Lipopolysaccharides/pharmacology , Membrane Lipids/metabolism , Mice , Microglia/cytology , Neuroimmunomodulation/physiology , Oxidation-Reduction , Oxidative Phosphorylation
19.
Mol Nutr Food Res ; 63(7): e1800813, 2019 04.
Article in English | MEDLINE | ID: mdl-30632684

ABSTRACT

SCOPE: The mechanisms and involvement of uncoupling protein 1 (UCP1) in the protection from obesity and insulin resistance induced by intake of a high-fat diet rich in omega-3 (n-3) fatty acids are investigated. METHODS AND RESULTS: C57BL/6J mice are fed either a low-fat (control group) or one of two isocaloric high-fat diets containing either lard (HFD) or fish oil (HFN3) as fat source and evaluated for body weight, adiposity, energy expenditure, glucose homeostasis, and inguinal white and interscapular brown adipose tissue (iWAT and iBAT, respectively) gene expression, lipidome, and mitochondrial bioenergetics. HFN3 intake protected from obesity, glucose and insulin intolerances, and hyperinsulinemia. This is associated with increased energy expenditure, iWAT UCP1 expression, and incorporation of n-3 eicosapentaenoic and docosahexaenoic fatty acids in iWAT and iBAT triacylglycerol. Importantly, HFN3 is equally effective in reducing body weight gain, adiposity, and glucose intolerance and increasing energy expenditure in wild-type and UCP1-deficient mice without recruiting other thermogenic processes in iWAT and iBAT, such as mitochondrial uncoupling and SERCA-mediated calcium and creatine-driven substrate cyclings. CONCLUSION: Intake of a high-fat diet rich in omega-3 fatty acids protects both wild-type and UCP1-deficient mice from obesity and insulin resistance by increasing energy expenditure through unknown mechanisms.


Subject(s)
Energy Metabolism/drug effects , Fish Oils/pharmacology , Glucose Intolerance/diet therapy , Obesity/prevention & control , Uncoupling Protein 1/genetics , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Diet, High-Fat/adverse effects , Energy Metabolism/genetics , Fatty Acids, Omega-3/analysis , Fatty Acids, Omega-3/pharmacology , Fish Oils/chemistry , Glucose Intolerance/genetics , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Thermogenesis/drug effects , Thermogenesis/genetics , Uncoupling Protein 1/metabolism
20.
Redox Biol ; 19: 105-115, 2018 10.
Article in English | MEDLINE | ID: mdl-30142602

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by degeneration of upper and lower motor neurons. While the fundamental causes of the disease are still unclear, the accumulation of Cu,Zn-superoxide dismutase (SOD1) immunoreactive aggregates is associated with familial ALS cases. Cholesterol 5,6-secosterol aldehydes (Seco A and Seco B) are reported to contribute to neurodegenerative disease pathology by inducing protein modification and aggregation. Here we have investigated the presence of secosterol aldehydes in ALS SOD1-G93A rats and their capacity to induce SOD1 aggregation. Secosterol aldehydes were analyzed in blood plasma, spinal cord and motor cortex of ALS rats at the pre-symptomatic and symptomatic stages. Seco B was significantly increased in plasma of symptomatic ALS rats compared to pre-symptomatic animals, suggesting an association with disease progression. In vitro experiments showed that both Seco A and Seco B induce the formation of high molecular weight (HMW) SOD1 aggregates with amorphous morphology. SOD1 adduction to ω-alkynyl-secosterols analyzed by click assay showed that modified proteins are only detected in the HMW region, indicating that secosterol adduction generates species highly prone to aggregate. Of note, SOD1-secosterol adducts containing up to five secosterol molecules were confirmed by MALDI-TOF analysis. Interestingly, mass spectrometry sequencing of SOD1 aggregates revealed preferential secosterol adduction to Lys residues located at the electrostatic loop (Lys 122, 128 and 136) and nearby the dimer interface (Lys 3 and 9). Altogether, our results show that secosterol aldehydes are increased in plasma of symptomatic ALS rats and represent a class of aldehydes that can potentially modify SOD1 enhancing its propensity to aggregate.


Subject(s)
Aldehydes/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Cholesterol/analogs & derivatives , Protein Aggregation, Pathological/metabolism , Superoxide Dismutase-1/metabolism , Aldehydes/blood , Amyotrophic Lateral Sclerosis/blood , Amyotrophic Lateral Sclerosis/genetics , Animals , Cholesterol/blood , Cholesterol/metabolism , Disease Models, Animal , Humans , Male , Point Mutation , Protein Aggregates , Protein Aggregation, Pathological/blood , Protein Aggregation, Pathological/genetics , Rats, Sprague-Dawley , Rats, Transgenic , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...