Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters











Publication year range
1.
Biomedicines ; 10(6)2022 Jun 03.
Article in English | MEDLINE | ID: mdl-35740338

ABSTRACT

Exosomes/microvesicles originate from multivesicular bodies that allow the secretion of endolysosome components out of the cell. In the present work, we investigated the effects of rottlerin, a polyphenol, on exosome/microvesicle secretion in a model of intracellular lipid trafficking impairment, and elucidated the mechanism of action. In a model of lipid trafficking impairment in C6 glia cells, rottlerin increased ceramide levels, while decreasing hexosylceramide content. This was accompanied by increased exosome/microvesicle secretion, thereby reducing the concentration of lipids in the endolysosomal compartment. The reduction of hexosylceramide levels by rottlerin was attributed to the increase of ß-glucosidase (glucosylceramidase) activity, and the effects of rottlerin were abrogated by ß-glucosidase inhibitors such as isofagomine D-tartrate and AMP-deoxynojirimycin. Moreover, treatment with ML-266, a potent activator of the ß-glucosidase enzyme, recapitulated the effects of rottlerin on the sphingolipid profile and exosome/microvesicle secretion. Finally, inhibition of AMPK (AMP-activated protein kinase) using compound C prevented both exosome/microvesicle secretion and the elimination of endolysosome lipids, which were promoted by rottlerin. The results showed that the decrease in intracellular lipid deposition induced by rottlerin was mediated by ß-glucosidase activation and exosome/microvesicle release via the AMPK pathway. Rottlerin consumption could represent an additional health benefit in lysosomal deposition diseases.

2.
Int J Mol Sci ; 23(8)2022 Apr 10.
Article in English | MEDLINE | ID: mdl-35456988

ABSTRACT

Squalene is a natural bioactive triterpene and an important intermediate in the biosynthesis of sterols. To assess the effect of this compound on the hepatic transcriptome, RNA-sequencing was carried out in two groups of male New Zealand rabbits fed either a diet enriched with 1% sunflower oil or the same diet with 0.5% squalene for 4 weeks. Hepatic lipids, lipid droplet area, squalene, and sterols were also monitored. The Squalene administration downregulated 9 transcripts and upregulated 13 transcripts. The gene ontology of transcripts fitted into the following main categories: transporter of proteins and sterols, lipid metabolism, lipogenesis, anti-inflammatory and anti-cancer properties. When the results were confirmed by RT-qPCR, rabbits receiving squalene displayed significant hepatic expression changes of LOC100344884 (PNPLA3), GCK, TFCP2L1, ASCL1, ACSS2, OST4, FAM91A1, MYH6, LRRC39, LOC108176846, GLT1D1 and TREH. A squalene-enriched diet increased hepatic levels of squalene, lanosterol, dihydrolanosterol, lathosterol, zymostenol and desmosterol. Strong correlations were found among specific sterols and some squalene-changed transcripts. Incubation of the murine AML12 hepatic cell line in the presence of lanosterol, dihydrolanosterol, zymostenol and desmosterol reproduced the observed changes in the expressions of Acss2, Fam91a1 and Pnpla3. In conclusion, these findings indicate that the squalene and post-squalene metabolites play important roles in hepatic transcriptional changes required to protect the liver against malfunction.


Subject(s)
Lanosterol , Squalene , Acyltransferases , Animals , Desmosterol/metabolism , Desmosterol/pharmacology , Lanosterol/pharmacology , Liver/metabolism , Male , Mice , Phospholipases A2, Calcium-Independent/metabolism , Rabbits , Squalene/pharmacology , Sterols/metabolism , Transcriptome
3.
Biochem Pharmacol ; 196: 114623, 2022 02.
Article in English | MEDLINE | ID: mdl-34052188

ABSTRACT

The mevalonate pathway is responsible for the synthesis of isoprenoids, including sterols and other metabolites that are essential for diverse biological functions. Cholesterol, the main sterol in mammals, and non-sterol isoprenoids are in high demand by rapidly dividing cells. As evidence of its importance, many cell signaling pathways converge on the mevalonate pathway and these include those involved in proliferation, tumor-promotion, and tumor-suppression. As well as being a fundamental building block of cell membranes, cholesterol plays a key role in maintaining their lipid organization and biophysical properties, and it is crucial for the function of proteins located in the plasma membrane. Importantly, cholesterol and other mevalonate derivatives are essential for cell cycle progression, and their deficiency blocks different steps in the cycle. Furthermore, the accumulation of non-isoprenoid mevalonate derivatives can cause DNA replication stress. Identification of the mechanisms underlying the effects of cholesterol and other mevalonate derivatives on cell cycle progression may be useful in the search for new inhibitors, or the repurposing of preexisting cholesterol biosynthesis inhibitors to target cancer cell division. In this review, we discuss the dependence of cell division on an active mevalonate pathway and the role of different mevalonate derivatives in cell cycle progression.


Subject(s)
Cell Cycle/physiology , Cholesterol/metabolism , Mevalonic Acid/metabolism , Sterols/metabolism , Terpenes/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Signal Transduction/drug effects , Signal Transduction/physiology
4.
Nat Commun ; 12(1): 6448, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34750386

ABSTRACT

Intricate regulatory networks govern the net balance of cholesterol biosynthesis, uptake and efflux; however, the mechanisms surrounding cholesterol homeostasis remain incompletely understood. Here, we develop an integrative genomic strategy to detect regulators of LDLR activity and identify 250 genes whose knockdown affects LDL-cholesterol uptake and whose expression is modulated by intracellular cholesterol levels in human hepatic cells. From these hits, we focus on MMAB, an enzyme which catalyzes the conversion of vitamin B12 to adenosylcobalamin, and whose expression has previously been linked with altered levels of circulating cholesterol in humans. We demonstrate that hepatic levels of MMAB are modulated by dietary and cellular cholesterol levels through SREBP2, the master transcriptional regulator of cholesterol homeostasis. Knockdown of MMAB decreases intracellular cholesterol levels and augments SREBP2-mediated gene expression and LDL-cholesterol uptake in human and mouse hepatic cell lines. Reductions in total sterol content were attributed to increased intracellular levels of propionic and methylmalonic acid and subsequent inhibition of HMGCR activity and cholesterol biosynthesis. Moreover, mice treated with antisense inhibitors of MMAB display a significant reduction in hepatic HMGCR activity, hepatic sterol content and increased expression of SREBP2-mediated genes. Collectively, these findings reveal an unexpected role for the adenosylcobalamin pathway in regulating LDLR expression and identify MMAB as an additional control point by which cholesterol biosynthesis is regulated by its end product.


Subject(s)
Cholesterol/metabolism , Feedback, Physiological , Homeostasis , Liver/metabolism , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , Animals , Cell Line, Tumor , Cholesterol, LDL/metabolism , Gene Expression Profiling/methods , HeLa Cells , Hep G2 Cells , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic/genetics , RNA Interference , Receptors, LDL/genetics , Receptors, LDL/metabolism , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism
5.
Food Funct ; 12(17): 8141-8153, 2021 Sep 07.
Article in English | MEDLINE | ID: mdl-34291245

ABSTRACT

To evaluate the effects of squalene, the main unsaponifiable component of virgin olive oil, on lipid metabolism, two groups of male New Zealand rabbits were fed a 1% sunflower oil-enriched regular diet or the same diet containing 0.5% squalene for 4 weeks. Plasma triglycerides, total- and HDL-cholesterol and their lipoproteins were assayed. Analyses of hepatic lipid droplets, triglycerides, total- and non-esterified cholesterol, squalene, protein and gene expression, and cholesterol precursors were carried out. In the jejunum, the squalene content and mRNA and protein APOB expressions were measured. Finally, we studied the effect of cholesterol precursors in AML12 cells. Squalene administration significantly increased plasma total cholesterol, mainly carried as non-esterified cholesterol in IDL and large LDL, and corresponded to an increased number of APOB100-containing particles without accumulation of triglycerides and decreased reactive oxygen species. Despite no significant changes in the APOB content in the jejunum, the latter displayed increased APOB mRNA and squalene levels. Increases in the amounts of non-esterified cholesterol, squalene, lanosterol, dihydrolanosterol, lathosterol, cholestanol, zymostenol, desmosterol and caspase 1 were also observed in the liver. Incubation of AML12 cells in the presence of lanosterol increased caspase 1. In conclusion, squalene administration in rabbits increases the number of modified APOB-containing lipoproteins, and hepatic cholesterol biosynthesis is linked to caspase 1 probably through lanosterol.


Subject(s)
Cholesterol/metabolism , Hypercholesterolemia/diet therapy , Lipoproteins/blood , Liver/metabolism , Squalene/metabolism , Animals , Apolipoproteins B/genetics , Apolipoproteins B/metabolism , Cholesterol/blood , Cholesterol, HDL/blood , Humans , Hypercholesterolemia/blood , Male , Rabbits , Triglycerides/blood
6.
Biomed Pharmacother ; 141: 111871, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34225017

ABSTRACT

Selective estrogen receptor modulators (SERMs) are nonsteroidal drugs that display an estrogen-agonist or estrogen-antagonist effect depending on the tissue targeted. SERMs have attracted great clinical interest for the treatment of several pathologies, most notably breast cancer and osteoporosis. There is strong evidence that SERMs secondarily affect cholesterol metabolism, although the mechanism has not been fully elucidated. In this study, we analysed the effect of the SERMs tamoxifen, raloxifene, and toremifene on the expression of lipid metabolism genes by microarrays and quantitative PCR in different cell types, and ascertained the main mechanisms involved. The three SERMs increased the expression of sterol regulatory element-binding protein (SREBP) target genes, especially those targeted by SREBP-2. In consonance, SERMs increased SREBP-2 processing. These effects were associated to the interference with intracellular LDL-derived cholesterol trafficking. When the cells were exposed to LDL, but not to cholesterol/methyl-cyclodextrin complexes, the SERM-induced increases in gene expression were synergistic with those induced by lovastatin. Furthermore, the SERMs reduced the stimulation of the transcriptional activity of the liver X receptor (LXR) by exogenous cholesterol. However, their impact on the expression of the LXR canonical target ABCA1 in the presence of LDL was cell-type dependent. These actions of SERMs were independent of estrogen receptors. We conclude that, by inhibiting the intracellular trafficking of LDL-derived cholesterol, SERMs promote the activation of SREBP-2 and prevent the activation of LXR, two master regulators of cellular cholesterol metabolism. This study highlights the impact of SERMs on lipid homeostasis regulation beyond their actions as estrogen receptor modulators.


Subject(s)
Cholesterol/metabolism , Homeostasis/drug effects , Liver X Receptors/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Sterol Regulatory Element Binding Protein 2/metabolism , Cholesterol, LDL/metabolism , Hep G2 Cells , Homeostasis/physiology , Humans , Liver X Receptors/antagonists & inhibitors , MCF-7 Cells
7.
Int J Mol Sci ; 22(8)2021 Apr 10.
Article in English | MEDLINE | ID: mdl-33920193

ABSTRACT

Atypical or second-generation antipsychotics are used in the treatment of psychosis and behavioral problems in older persons with dementia. However, these pharmaceutical drugs are associated with an increased risk of stroke in such patients. In this study, we evaluated the effects of risperidone treatment on phospholipid and sphingolipid composition and lipid raft function in peripheral blood mononuclear cells (PBMCs) of older patients (mean age >88 years). The results showed that the levels of dihydroceramides, very-long-chain ceramides, and lysophosphatidylcholines decreased in PBMCs of the risperidone-treated group compared with untreated controls. These findings were confirmed by in vitro assays using human THP-1 monocytes. The reduction in the levels of very-long-chain ceramides and dihydroceramides could be due to the decrease in the expression of fatty acid elongase 3, as observed in THP-1 monocytes. Moreover, risperidone disrupted lipid raft domains in the plasma membrane of PBMCs. These results indicated that risperidone alters phospholipid and sphingolipid composition and lipid raft domains in PBMCs of older patients, potentially affecting multiple signaling pathways associated with these membrane domains.


Subject(s)
Ceramides/metabolism , Lipid Metabolism/drug effects , Psychotic Disorders/drug therapy , Aged , Aged, 80 and over , Antipsychotic Agents/pharmacology , Cell Membrane/genetics , Cell Membrane/metabolism , Female , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lipid Metabolism/genetics , Lysophospholipids/genetics , Male , Olanzapine/pharmacology , Psychotic Disorders/blood , Psychotic Disorders/pathology , Risperidone/pharmacology , Sphingolipids/genetics
8.
Semin Cancer Biol ; 73: 101-115, 2021 08.
Article in English | MEDLINE | ID: mdl-32931953

ABSTRACT

Selective estrogen receptor modulators (SERMs) are a class of compounds that bind to estrogen receptors (ERs) and possess estrogen agonist or antagonist actions in different tissues. As such, they are widely used drugs. For instance, tamoxifen, the most prescribed SERM, is used to treat ERα-positive breast cancer. Aside from their therapeutic targets, SERMs have the capacity to broadly affect cellular cholesterol metabolism and handling, mainly through ER-independent mechanisms. Cholesterol metabolism reprogramming is crucial to meet the needs of cancer cells, and different key processes involved in cholesterol homeostasis have been associated with cancer progression. Therefore, the effects of SERMs on cholesterol homeostasis may be relevant to carcinogenesis, either by contributing to the anticancer efficacy of these compounds or, conversely, by promoting resistance to treatment. Understanding these aspects of SERMs actions could help to design more efficacious therapies. Herein we review the effects of SERMs on cellular cholesterol metabolism and handling and discuss their potential in anticancer pharmacology.


Subject(s)
Cholesterol/metabolism , Lipid Metabolism/drug effects , Neoplasms , Selective Estrogen Receptor Modulators/pharmacology , Animals , Humans , Lipid Metabolism/physiology , Neoplasms/drug therapy , Neoplasms/metabolism
9.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1865(12): 158790, 2020 12.
Article in English | MEDLINE | ID: mdl-32771460

ABSTRACT

BACKGROUND AND AIMS: The molecular mechanisms by which the liver develops steatotic disease still remain unclear. Previous studies using nutritional and genetic models of hepatic steatosis in mice showed that liver synaptotagmin 1 (Syt1) expression was associated with lipid droplet area. Hepatic Syt1 overexpression was used as a tool to explore its effect on hepatic and plasma lipids. METHODS AND RESULTS: To find out a cause-effect, hepatic mouse Syt1 mRNA was cloned into a vector driving hepatocyte-specific expression and administered by hydrodynamic injection to male Apoe-deficient mice fed on a Western diet, the latter as a model of rapid spontaneous steatosis development. Hepatic microsomal, large vesicle, lysosomal and plasma membrane fractions were enriched in SYT1 protein following gene overexpression. In these conditions, very low density lipoprotein esterified cholesterol increased. Likewise, the transgene caused an alteration in lipid droplet surface and a positive correlation between Syt1 expression and hepatic total cholesterol content. A lipidomic approach evidenced a decrease in lysophosphatidylcholine, phosphatidylcholine and triglycerides in isolated plasma membrane fraction. Expressions of genes involved in biosynthesis of bile acids, fatty acid metabolism, lipoprotein dynamics and vesicular transport were modified by the increased SYT1 expression. CONCLUSIONS: These results indicate that this protein is involved in hepatic management of lipids and in the regulation of genes involved in lipid metabolism.


Subject(s)
Apolipoproteins E/genetics , Diet, Western , Lipid Metabolism , Liver/metabolism , Synaptotagmin I/metabolism , Animals , Apolipoproteins E/metabolism , Cell Membrane/genetics , Cell Membrane/metabolism , Diet, Western/adverse effects , Fatty Liver/etiology , Fatty Liver/genetics , Fatty Liver/metabolism , Gene Deletion , Gene Expression , Hep G2 Cells , Humans , Lipid Droplets/metabolism , Male , Mice , Mice, Inbred C57BL , Synaptotagmin I/genetics
10.
Obes Facts ; 13(3): 321-330, 2020.
Article in English | MEDLINE | ID: mdl-32388504

ABSTRACT

BACKGROUND/AIM: Soluble tumor necrosis factor-like weak inducer of apoptosis (sTWEAK) and amyloid-ß(1-40) (Aß40) emerged as markers of cardiovascular risk because of their actions in the endothelium and their role in atherosclerotic progression. The aim of this study was to analyze the association of these two factors with the decrease in carotid intima-media thickness (cIMT) after bariatric surgery in obese women. METHODS: We studied 60 severely obese women, of whom 20 were submitted to laparoscopic Roux-en-Y gastric bypass (RYGB), 20 to sleeve gastrectomy (SG), and 20 to lifestyle modification therapy. Circulating sTWEAK, Aß40, high-sensitivity C-reactive protein, plasminogen activator inhibitor type 1, insulin resistance (HOMA-IR), and cIMT were measured at baseline and after 1 year of follow-up. RESULTS: sTWEAK increased similarly after both surgical procedures, whereas the increase observed after lifestyle intervention did not reach statistical significance. Aß40 showed no differences between groups of women, nor did it change during follow-up. The decrease in cIMT at 12 months correlated with the decrease in body mass index (BMI) (r = 0.45; p < 0.001) and fasting insulin (r = 0.30; p = 0.038), and also with the increase in sTWEAK (r = -0.43; p = 0.002). Multivariate linear regression showed that only the changes in BMI (ß = 0.389; p = 0.005) and sTWEAK (ß = -0.358; p = 0.009) were associated with the decrease in cIMT (R2 = 0.313; F = 9.348; p < 0.001). CONCLUSIONS: One year after bariatric surgery, RYGB and SG induced a similar increase in circulating sTWEAK that occurred in parallel to the decrease observed in cIMT.


Subject(s)
Cytokine TWEAK/metabolism , Obesity/surgery , Adult , Amyloid beta-Peptides/metabolism , Atherosclerosis , Bariatric Surgery , Body Mass Index , Carotid Intima-Media Thickness , Female , Gastrectomy/methods , Gastric Bypass , Humans , Male , Middle Aged , Obesity/metabolism , Peptide Fragments/metabolism
11.
Article in English | MEDLINE | ID: mdl-31988047

ABSTRACT

Curcumin, a hydrophobic polyphenol found in the rhizome of Curcuma longa, has been shown to reduce intracellular lipid accumulation in mouse models of lysosomal storage diseases such as Niemann-Pick type C. Exosomes are small extracellular vesicles secreted by cells in response to changes in intracellular ceramide composition. Curcumin can induce exosome/microvesicle release in cellular models of lipid deposition; however, the mechanism by which curcumin stimulates this release is unknown. In a model of lipid trafficking impairment in C6 glia cells, we show that curcumin stimulated ceramide synthesis by increasing the intracellular concentration of ceramide-dihydroceramide. Ceramide overload increased exosome/microvesicle secretion 10-fold, thereby reducing the concentration of lipids in the endolysosomal compartment. These effects were blocked by inhibitors of serine palmitoyltransferase (myriocin) and ceramide synthase (fumonisin B1). It is concluded that the decrease in intracellular lipid deposition induced by curcumin is mediated by increased ceramide synthesis and exosome/microvesicle release. This action may represent an additional health benefit of curcumin.


Subject(s)
Cell-Derived Microparticles/drug effects , Ceramides/biosynthesis , Curcumin/pharmacology , Exosomes/drug effects , Neuroglia/drug effects , Animals , Cell Line, Tumor , Cell-Derived Microparticles/metabolism , Exosomes/metabolism , Fatty Acids, Monounsaturated/pharmacology , Fumonisins/pharmacology , Humans , Lipid Metabolism/drug effects , Lipoproteins, LDL/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neuroglia/pathology , Niemann-Pick Disease, Type C/diet therapy , Niemann-Pick Disease, Type C/pathology , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/metabolism , Rats , Serine C-Palmitoyltransferase/antagonists & inhibitors , Serine C-Palmitoyltransferase/metabolism
12.
Circulation ; 140(3): 225-239, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31154825

ABSTRACT

BACKGROUND: Atherosclerosis is driven by synergistic interactions between pathological, biomechanical, inflammatory, and lipid metabolic factors. Our previous studies demonstrated that absence of caveolin-1 (Cav1)/caveolae in hyperlipidemic mice strongly inhibits atherosclerosis, which was attributed to activation of endothelial nitric oxide (NO) synthase (eNOS) and increased production of NO and reduced inflammation and low-density lipoprotein trafficking. However, the contribution of eNOS activation and NO production in the athero-protection of Cav1 and the exact mechanisms by which Cav1/caveolae control the pathogenesis of diet-induced atherosclerosis are still not clear. METHODS: Triple-knockout mouse lacking expression of eNOS, Cav1, and Ldlr were generated to explore the role of NO production in Cav1-dependent athero-protective function. The effects of Cav1 on lipid trafficking, extracellular matrix remodeling, and vascular inflammation were studied both in vitro and in vivo with a mouse model of diet-induced atherosclerosis. The expression of Cav1 and distribution of caveolae regulated by flow were analyzed by immunofluorescence staining and transmission electron microscopy. RESULTS: We found that absence of Cav1 significantly suppressed atherogenesis in Ldlr-/-eNOS-/- mice, demonstrating that athero-suppression is independent of increased NO production. Instead, we find that the absence of Cav1/caveolae inhibited low-density lipoprotein transport across the endothelium and proatherogenic fibronectin deposition and disturbed flow-mediated endothelial cell inflammation. Consistent with the idea that Cav1/caveolae may play a role in early flow-dependent inflammatory priming, distinct patterns of Cav1 expression and caveolae distribution were observed in athero-prone and athero-resistant areas of the aortic arch even in wild-type mice. CONCLUSIONS: These findings support a role for Cav1/caveolae as a central regulator of atherosclerosis that links biomechanical, metabolic, and inflammatory pathways independently of endothelial eNOS activation and NO production.


Subject(s)
Atherosclerosis/metabolism , Caveolin 1/physiology , Endothelium, Vascular/metabolism , Lipoproteins, LDL/metabolism , Nitric Oxide Synthase Type III/metabolism , Transcytosis/physiology , Animals , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Cells, Cultured , Dogs , Endothelium, Vascular/pathology , Enzyme Activation/physiology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
13.
Nutrients ; 11(6)2019 Jun 13.
Article in English | MEDLINE | ID: mdl-31200481

ABSTRACT

Postprandial lipemia has many physiopathological effects, some of which increase the risk of cardiovascular disease. MicroRNAs (miRNAs) can be found in almost all biological fluids, but their postprandial kinetics are poorly described. We aimed to profile circulating miRNAs in response to a fat challenge. In total, 641 circulating miRNAs were assessed by real-time PCR in plasmas from mice two hours after lipid gavage. Mice with intestine-specific loss of Dicer were screened to identify potential miRNAs released by the intestine. A total of 68 miRNAs were selected for further validation. Ten circulating miRNAs were finally validated as responsive to postprandial lipemia, including miR-206-3p, miR-543-3p, miR-466c-5p, miR-27b-5p, miR-409-3p, miR-340-3p, miR-1941-3p, miR-10a-3p, miR-125a-3p, and miR-468-3p. Analysis of their possible tissues of origin/target showed an enrichment of selected miRNAs in liver, intestine, brain, or skeletal muscle. miR-206, miR-27b-5p, and miR-409-3p were validated in healthy humans. Analysis of their predicted target genes revealed their potential involvement in insulin/insulin like growth factor (insulin/IGF), angiogenesis, cholecystokinin B receptor signaling pathway (CCKR), inflammation or Wnt pathways for mice, and in platelet derived growth factor (PDGF) and CCKR signaling pathways for humans. Therefore, the current study shows that certain miRNAs are released in the circulation in response to fatty meals, proposing them as potential novel therapeutic targets of lipid metabolism.


Subject(s)
Circulating MicroRNA/blood , Dietary Fats/adverse effects , Hyperlipidemias/etiology , Postprandial Period/drug effects , Animals , Humans , Mice , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects
14.
Atherosclerosis ; 284: 223-229, 2019 05.
Article in English | MEDLINE | ID: mdl-30777337

ABSTRACT

BACKGROUND AND AIMS: Autosomal recessive hypercholesterolemia (ARH) is a rare disorder caused by mutations in LDLRAP1, which impairs internalization of hepatic LDL receptor (LDLR). ARH patients respond relatively well to statins or the combination of statins and Ezetimibe, but scarce and variable data on treatment with PCSK9 inhibitors is available. We aimed to identify and characterize the defect in a hypercholesterolemic patient with premature cardiovascular disease and determine the response to lipid-lowering treatment. METHODS AND RESULTS: Gene sequencing revealed a homozygous c.1A > G:p.? variant in LDLRAP1. Primary lymphocytes were isolated from the ARH patient, one control and two LDLR-defective subjects, one LDLR:p.(Cys352Ser) heterozygote and one LDLR:p.(Asn825Lys) homozygote. The patient had undetectable full-length ARH protein by Western blotting, but expressed a lower-than-normal molecular weight peptide. LDLR activity was measured by flow cytometry, which showed that LDL binding and uptake were reduced in lymphocytes from the ARH patient as compared to control lymphocytes, but were slightly higher than in those from the LDLR:p.(Cys352Ser) heterozygote. Despite the analogous internalization defect predicted in ARH and homozygous LDLR:p.(Asn825Lys) lymphocytes, LDL uptake was higher in the former than in the latter. LDL-cholesterol levels were markedly reduced by the successive therapy with Atorvastatin and Atorvastatin plus Ezetimibe, and the addition of Evolocumab biweekly decreased LDL-cholesterol by a further 39%. CONCLUSIONS: The LDLRAP1:c.1A > G variant is associated with the appearance of an N-terminal truncated ARH protein and to reduced, although still significant, LDLR activity in lymphocytes. Residual LDLR activity may be relevant for the substantial response of the patient to Evolocumab.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Antibodies, Monoclonal, Humanized/therapeutic use , Anticholesteremic Agents/therapeutic use , Hypercholesterolemia/drug therapy , Hypercholesterolemia/genetics , Mutation , PCSK9 Inhibitors , Humans , Male , Middle Aged , Hyperlipoproteinemia Type III
15.
FASEB J ; 33(3): 3912-3921, 2019 03.
Article in English | MEDLINE | ID: mdl-30496704

ABSTRACT

Atherosclerosis is a chronic disease characterized by vascular lipid retention and inflammation, and pattern recognition receptors (PRRs) are important contributors in early stages of the disease. Given the implication of the intracellular PRR nucleotide-binding oligomerization domain 1 (NOD1) in cardiovascular diseases, we investigated its contribution to early atherosclerosis. We evidenced NOD1 induction in atherosclerotic human and mouse tissues, predominantly in vascular endothelial cells. Accordingly, NOD1 genetic inactivation in Apoe-/- mice reduced not only atherosclerosis burden, but also monocyte and neutrophil accumulation in atheromata. Of note, in the presence of either peptidoglycan or oxidized LDLs, endothelial NOD1 triggered VCAM-1 up-regulation through the RIP2-NF-κB axis in an autocrine manner, enhancing firm adhesion of both sets of myeloid cells to the inflamed micro- and macrovasculature in vivo. Our data define a major proatherogenic role for endothelial NOD1 in early leukocyte recruitment to the athero-prone vasculature, thus introducing NOD1 as an innovative therapeutic target and potential prognostic molecule.-González-Ramos, S., Paz-García, M., Rius, C., del Monte-Monge, A., Rodríguez, C., Fernández-García, V., Andrés, V., Martínez-González, J., Lasunción, M. A., Martín-Sanz, P., Soehnlein, O., Boscá, L. Endothelial NOD1 directs myeloid cell recruitment in atherosclerosis through VCAM-1.


Subject(s)
Atherosclerosis/metabolism , Cell Movement , Endothelium, Vascular/metabolism , Myeloid Cells/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Autocrine Communication , Cells, Cultured , Humans , Lipoproteins, LDL/metabolism , Male , Mice , Mice, Inbred C57BL , Myeloid Cells/physiology , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism
16.
J Nutr Biochem ; 63: 197-205, 2019 01.
Article in English | MEDLINE | ID: mdl-30414540

ABSTRACT

The evaluation of the benefits of omega-3 fatty acid supplementation in humans requires the identification and characterization of suitable biomarkers of its incorporation in the body. The reference method for the evaluation of omega-3, gas chromatography, is difficult to apply in clinical practice because of its low throughput and does not provide information about the incorporation of specific fatty acids in lipid species and the potential effects of supplementation on lipid classes. We used a quantitative lipidomic approach to follow the incorporation of omega-3 fatty acids into plasma lipids in cystic fibrosis patients (n=50) from a randomized controlled clinical trial after the supplementation of seaweed oil enriched with docosahexaenoic acid (DHA). Lipidomic analysis accurately showed the distribution of fatty acids in different lipid classes after omega-3 supplementation, and the performance in determining the compliance to supplementation was similar to that of gas chromatography coupled to mass spectrometry. Twelve months after fatty acid supplementation, DHA was predominantly incorporated into highly unsaturated cholesteryl esters (110.9±16.2 vs. 278.6±32.6 µM, mean±S.E.M.) and phosphatidylcholine (142.4±11.9 vs. 272.9±21.4 µM) and, to a lesser extent, into phosphatidylethanolamine (9.4±0.8 vs. 15.5±1.5 µM) and triglycerides (0.4±0.04 vs. 1.1±0.12 µM). In addition, a technique was developed for the fast measurement of the DHA/arachidonic acid ratio to simplify the follow-up of nutritional intervention with DHA-enriched foods. We conclude that lipidomics is a suitable approach for monitoring the incorporation of omega-3 fatty acids in nutritional studies.


Subject(s)
Cystic Fibrosis/diet therapy , Fatty Acids, Omega-3/pharmacology , Lipids/blood , Cystic Fibrosis/blood , Dietary Supplements , Docosahexaenoic Acids/pharmacology , Double-Blind Method , Fatty Acids/blood , Humans , Lipidomics/methods , Seaweed , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
17.
Nutrients ; 10(9)2018 Sep 07.
Article in English | MEDLINE | ID: mdl-30205424

ABSTRACT

The relationship between C-reactive protein (CRP) levels and plasma antioxidants has been established in adults. However, the association has been rarely investigated in healthy children. Thus, we examined the cross-sectional association of high-sensitivity CRP (hs-CRP) levels with fat-soluble plasma antioxidant concentrations in a cohort of healthy prepubertal children. We determined hs-CRP levels in 543 healthy six⁻eight-year-old children using a high-sensitivity CRP enzyme-linked immuno sorbent assay (ELISA) kit. The plasma concentrations of lipids, apolipoproteins and lipid-soluble antioxidants (α-tocopherol, γ-tocopherol, lycopene, α-carotene, ß-carotene and retinol) were determined using standardized methods. Pearson correlation analysis showed significant correlations between plasma hs-CRP and α-carotene and retinol concentrations. After adjusting by sex, body mass index (BMI) and lipid levels, only the association with retinol remains significant, with children in the highest hs-CRP tertile group (hs-CRP ≥ 0.60 mg/dL) showing significantly lower levels of retinol than those from the tertiles 1 and 2. A stepwise linear regression selected retinol, BMI, apo A-I and sex as predictors of hs-CRP levels, in a model explaining 19.2% of the variability of hs-CRP. In conclusion, in healthy prepubertal children, after adjusting by sex, BMI and lipid levels, hs-CRP concentrations were highly associated with plasma retinol, which is transported in blood bound to retinol-binding protein but were not associated with the lipoprotein-bound antioxidants.


Subject(s)
Antioxidants/analysis , C-Reactive Protein/analysis , Inflammation Mediators/blood , Vitamin A/blood , Age Factors , Biomarkers/blood , Child , Cross-Sectional Studies , Female , Humans , Male , Spain
18.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(9): 958-967, 2018 09.
Article in English | MEDLINE | ID: mdl-29793057

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE), the most common model for multiple sclerosis, is characterized by inflammatory cell infiltration into the central nervous system and demyelination. Previous studies have demonstrated that administration of some polyphenols may reduce the neurological alterations of EAE. In this work, we show that ellagic acid, a polyphenolic compound, is beneficial in EAE, most likely through stimulation of ceramide biosynthesis within the brain. EAE was induced in Lewis rats by injection of guinea-pig spinal cord tissue along with Freund's complete adjuvant containing Mycobacterium tuberculosis. Clinical signs first appeared at day 8 post-immunization and reached a peak within 3 days, coincident with reduction of myelin basic protein (MBP) in the cortex. Sphingolipids, the other major components of myelin, also decreased at the acute phase of EAE, both in the cerebral cortex and in the spinal cord. In rats receiving ellagic acid in the drinking water from 2 days before immunization, the onset of the disease was delayed and clinical signs were reduced. This amelioration of clinical signs was accompanied by sustained levels of both MBP and sphingolipid in the cortex, without apparent changes in infiltration of inflammatory CD3+ T-cells, microglial activation, or weight loss, which together suggest a neuroprotective effect of ellagic acid. Finally, in glioma and oligodendroglioma cells we demonstrate that urolithins, the ellagic acid metabolites that circulate in plasma, stimulate the synthesis of ceramide. Together these data suggest that ellagic acid consumption protects against demyelination in rats with induced EAE, likely by a mechanism involving sphingolipid synthesis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Ceramides/agonists , Ellagic Acid/pharmacology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Myelin Sheath/drug effects , Neuroprotective Agents/pharmacology , Animals , Cell Line, Tumor , Ceramides/biosynthesis , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Coumarins/metabolism , Coumarins/pharmacology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Freund's Adjuvant/administration & dosage , Gene Expression , Guinea Pigs , Mycobacterium tuberculosis/chemistry , Myelin Basic Protein/agonists , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Myelin Sheath/metabolism , Myelin Sheath/pathology , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/pathology , Rats , Rats, Inbred Lew , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology
19.
Cell Rep ; 19(13): 2743-2755, 2017 06 27.
Article in English | MEDLINE | ID: mdl-28658622

ABSTRACT

Macrophages perform critical functions in both innate immunity and cholesterol metabolism. Here, we report that activation of Toll-like receptor 4 (TLR4) in macrophages causes lanosterol, the first sterol intermediate in the cholesterol biosynthetic pathway, to accumulate. This effect is due to type I interferon (IFN)-dependent histone deacetylase 1 (HDAC1) transcriptional repression of lanosterol-14α-demethylase, the gene product of Cyp51A1. Lanosterol accumulation in macrophages, because of either treatment with ketoconazole or induced conditional disruption of Cyp51A1 in mouse macrophages in vitro, decreases IFNß-mediated signal transducer and activator of transcription (STAT)1-STAT2 activation and IFNß-stimulated gene expression. These effects translate into increased survival to endotoxemic shock by reducing cytokine secretion. In addition, lanosterol accumulation increases membrane fluidity and ROS production, thus potentiating phagocytosis and the ability to kill bacteria. This improves resistance of mice to Listeria monocytogenes infection by increasing bacterial clearance in the spleen and liver. Overall, our data indicate that lanosterol is an endogenous selective regulator of macrophage immunity.


Subject(s)
Lanosterol/immunology , Macrophages/immunology , Toll-Like Receptor 4/immunology , Animals , Down-Regulation , Female , Gene Knockout Techniques , Humans , Immunity, Innate/drug effects , Lanosterol/metabolism , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Sterol 14-Demethylase/immunology
20.
Sci Rep ; 6: 32105, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27601313

ABSTRACT

Selective estrogen receptor modulators (SERMs) are widely prescribed drugs that alter cellular and whole-body cholesterol homeostasis. Here we evaluate the effect of SERMs on the macrophage-specific reverse cholesterol transport (M-RCT) pathway, which is mediated by HDL. Treatment of human and mouse macrophages with tamoxifen, raloxifene or toremifene induced the accumulation of cytoplasmic vesicles of acetyl-LDL-derived free cholesterol. The SERMs impaired cholesterol efflux to apolipoprotein A-I and HDL, and lowered ABCA1 and ABCG1 expression. These effects were not altered by the antiestrogen ICI 182,780 nor were they reproduced by 17ß-estradiol. The treatment of mice with tamoxifen or raloxifene accelerated HDL-cholesteryl ester catabolism, thereby reducing HDL-cholesterol concentrations in serum. When [(3)H]cholesterol-loaded macrophages were injected into mice intraperitoneally, tamoxifen, but not raloxifene, decreased the [(3)H]cholesterol levels in serum, liver and feces. Both SERMs downregulated liver ABCG5 and ABCG8 protein expression, but tamoxifen reduced the capacity of HDL and plasma to promote macrophage cholesterol efflux to a greater extent than raloxifene. We conclude that SERMs interfere with intracellular cholesterol trafficking and efflux from macrophages. Tamoxifen, but not raloxifene, impair M-RCT in vivo. This effect is primarily attributable to the tamoxifen-mediated reduction of the capacity of HDL to promote cholesterol mobilization from macrophages.


Subject(s)
Cholesterol/metabolism , Lipoproteins, HDL/metabolism , Macrophages/drug effects , Selective Estrogen Receptor Modulators/pharmacology , ATP Binding Cassette Transporter 1/biosynthesis , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter, Subfamily G/biosynthesis , ATP Binding Cassette Transporter, Subfamily G/genetics , Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Animals , Apolipoprotein A-I/metabolism , Biological Transport/drug effects , Cholesterol/analysis , Cholesterol/blood , Cholesterol Esters/metabolism , Diet, Western , Esterification/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Feces/chemistry , Fulvestrant , Humans , Lipoproteins, LDL/metabolism , Liver/chemistry , Liver/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Raloxifene Hydrochloride/pharmacology , THP-1 Cells , Tamoxifen/pharmacology , Toremifene/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL