Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 180
Filter
1.
FASEB J ; 35(5): e21494, 2021 05.
Article in English | MEDLINE | ID: mdl-33856696

ABSTRACT

Monocyte recruitment after vascular injury and their migration through the vessel wall represent crucial events in the initiation, progression, and destabilization of atherosclerotic plaque. Circulating monocytes are exposed to stimuli that alter their physiological state, and among them, lipids play a key role. Several studies investigated the mechanisms by which lipids affect monocyte functions promoting coronary atherosclerotic plaque initiation, but information on the relationship between lipid composition and function of monocyte is scant. We aimed at studying the migration of circulating monocytes isolated from patients with acute myocardial infarction (AMI) at hospital presentation and investigating its correlation with cellular lipid profile. The migration of monocytes was tested using both fetal bovine serum (FBS) and autologous serum as chemoattractant stimuli. Monocyte lipid profile was evaluated through an untargeted lipidomics approach, using a liquid chromatography/time-of-flight mass spectrometry platform. We observed that AMI patients' monocytes showed a significant increase in FBS and autologous serum-mediated migration compared to controls. Moreover, a different monocyte lipidomic profile between the two study groups was detected. In particular, AMI patients' monocytes showed an altered composition in ceramides, with an increase in lactosylceramide and in phospholipids (ie, phosphatidylethanolamine and lisophosphatidylethanolamine). Of note, a positive correlation between lactosylceramide levels and monocyte migration was observed. Furthermore, the lactosylceramide synthase inhibition significantly reduced FBS-induced monocyte migration. Our results highlight the influence of lactosylceramide on the monocyte migration capacity, pointing out a new possible mechanism of lipids in the onset of atherothrombosis and, hence, in AMI.


Subject(s)
Cell Movement , Lactosylceramides/metabolism , Lipidomics/methods , Lipids/analysis , Monocytes/metabolism , Myocardial Infarction/pathology , Female , Humans , Male , Middle Aged , Myocardial Infarction/metabolism
2.
Glycoconj J ; 39(2): 239-246, 2022 04.
Article in English | MEDLINE | ID: mdl-35377103

ABSTRACT

The innate immune system of mammalian cells is the first line of defense against pathogenic microorganisms. Phagocytes, which play the central role in this system, engulf microorganisms by a mechanism that involves pattern recognition receptors on their own surface and pathogen-associated molecular patterns (PAMPs) expressed by the microorganism. Components of PAMPs include glycans (polysaccharides) and glycoconjugates (carbohydrates covalently linked to other biological molecules). Pathogenic microorganisms display specific binding affinity to various types of glycosphingolipids (sphingosine-containing glycolipids; GSLs), and GSLs are involved in host-pathogen interactions. We observed that lactosylceramide (LacCer), a neutral GSL, binds directly to certain pathogen-specific molecules (e.g., Candida albicans-derived ß-glucans, mycobacterial lipoarabinomannan) via carbohydrate-carbohydrate interaction. LacCer is expressed highly on human neutrophils, and forms membrane microdomains. Such LacCer-enriched microdomains mediate several important neutrophil functions, including chemotaxis, phagocytosis, and superoxide generation. Human neutrophils phagocytose pathogenic mycobacteria (including Mycobacterium tuberculosis) through carbohydrate-carbohydrate interaction between LacCer on their own surface and mannose-capped lipoarabinomannan on the bacterium. During recognition of pathogen-specific glycans, direct association of LacCer-containing C24 fatty acid chain with Lyn (a Src family kinase) is necessary for signal transduction from the neutrophil exterior to interior. Pathogenic mycobacteria utilize a similar interaction to avoid killing by neutrophils. We describe here the mechanisms whereby LacCer mediates neutrophil immune systems via carbohydrate-carbohydrate interaction.


Subject(s)
Mycobacterium , Neutrophils , Animals , Antigens, CD/metabolism , Glycosphingolipids/metabolism , Humans , Lactosylceramides/metabolism , Mammals/metabolism , Membrane Microdomains/metabolism , Mycobacterium/metabolism , Neutrophils/metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism
3.
Int J Mol Sci ; 23(23)2022 Dec 06.
Article in English | MEDLINE | ID: mdl-36499769

ABSTRACT

Diabetes contributes to about 30% morbidity and mortality world-wide and has tidal wave increases in several countries in Asia. Diabetes is a multi-factorial disease compounded by inflammation, dyslipidemia, atherosclerosis, and is sometimes accompanied with gains in body weight. Sphingolipid pathways that interplay in the enhancement of the pathology of this disease may be potential therapeutic targets. Thus, the application of advanced sphingolipidomics may help predict the progression of this disease and therapeutic outcomes in man. Pre-clinical studies using various experimental animal models of diabetes provide valuable information on the role of sphingolipid signaling networks in diabetes and the efficacy of drugs to determine the translatability of innovative discoveries to man. In this review, we discuss three major concepts regarding sphingolipids and diabetes. First, we discuss a possible involvement of a monosialodihexosylceramide (GM3) in insulin-insulin receptor interactions. Second, a potential role for ceramide (Cer) and lactosylceramide (LacCer) in apoptosis and mitochondrial dysfunction is proposed. Third, a larger role of LacCer in antioxidant status and inflammation is discussed. We also discuss how inhibitors of glycosphingolipid synthesis can ameliorate diabetes in experimental animal models.


Subject(s)
Cardiovascular Diseases , Diabetes Mellitus , Animals , Glycosphingolipids/metabolism , Cardiovascular Diseases/prevention & control , Sphingolipids/metabolism , Lactosylceramides/metabolism , Oxidative Stress , Inflammation , Models, Animal
4.
Hum Mol Genet ; 28(15): 2514-2530, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31070736

ABSTRACT

A rare lysosomal disease resembling a mucopolysaccharidosis with unusual systemic features, including renal disease and platelet dysfunction, caused by the defect in a conserved region of the VPS33A gene on human chromosome 12q24.31, occurs in Yakuts-a nomadic Turkic ethnic group of Southern Siberia. VPS33A is a core component of the class C core vacuole/endosome tethering (CORVET) and the homotypic fusion and protein sorting (HOPS) complexes, which have essential functions in the endocytic pathway. Here we show that cultured fibroblasts from patients with this disorder have morphological changes: vacuolation with disordered endosomal/lysosomal compartments and-common to sphingolipid diseases-abnormal endocytic trafficking of lactosylceramide. Urine glycosaminoglycan studies revealed a pathological excess of sialylated conjugates as well as dermatan and heparan sulphate. Lipidomic screening showed elevated ß-D-galactosylsphingosine with unimpaired activity of cognate lysosomal hydrolases. The 3D crystal structure of human VPS33A predicts that replacement of arginine 498 by tryptophan will de-stabilize VPS33A folding. We observed that the missense mutation reduced the abundance of full-length VPS33A and other components of the HOPS and CORVET complexes. Treatment of HeLa cells stably expressing the mutant VPS33A with a proteasome inhibitor rescued the mutant protein from degradation. We propose that the disease is due to diminished intracellular abundance of intact VPS33A. Exposure of patient-derived fibroblasts to the clinically approved proteasome inhibitor, bortezomib, or inhibition of glucosylceramide synthesis with eliglustat, partially corrected the impaired lactosylceramide trafficking defect and immediately suggest therapeutic avenues to explore in this fatal orphan disease.


Subject(s)
Antigens, CD/metabolism , Carbohydrate Metabolism, Inborn Errors/genetics , Endocytosis , Lactosylceramides/metabolism , Lysosomes/metabolism , Mutation, Missense , Vesicular Transport Proteins/genetics , Bortezomib/therapeutic use , Carbohydrate Metabolism, Inborn Errors/metabolism , Carbohydrate Metabolism, Inborn Errors/physiopathology , Cells, Cultured , Female , Fibroblasts/metabolism , Fibroblasts/pathology , HeLa Cells , Humans , Infant , Lysosomes/physiology , Male , Mucopolysaccharidoses , Phenotype , Proteasome Inhibitors/therapeutic use , Protein Conformation , Pyrrolidines/therapeutic use , Siberia , Vesicular Transport Proteins/metabolism , Exome Sequencing
5.
Med Sci Monit ; 27: e930166, 2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33790218

ABSTRACT

BACKGROUND Fluorofenidone (AKF-PD) is an anti-fibrotic small-molecule compound. Its mechanism of action on paraquat (PQ)-induced pulmonary fibrosis is still unclear. MATERIAL AND METHODS Forty-eight SD rats were divided into 4 groups: control group, PQ group, PQ+AKF-PD group, and AKF-PD group. The pathological changes of lung tissues were observed by Masson and HE staining. The UPLC-QTOF-MS analysis was performed to detect the differences in metabolites among groups, then the possible mechanisms of the anti-pulmonary fibrosis effects of fluorofenidone were further revealed by network pharmacology analysis. Biological methods were used to verify the results of the network pharmacology analysis. RESULTS The results showed that fluorofenidone treatment significantly alleviated paraquat-induced pulmonary fibrosis. Metabolomics analysis showed that 18 metabolites were disordered in the serum of paraquat-poisoned rats, of which 13 were restored following fluorofenidone treatment. Network pharmacology analysis showed that the drug screened a total of 12 targets and mainly involved multiple signaling pathways and metabolic pathways to jointly exert anti-pulmonary fibrosis effects. Autophagy is the main pathway of fluorofenidone in treatment pulmonary fibrosis. The western blot results showed that fluorofenidone upregulated the expression of LC3-II/I and E-cadherin, and downregulated the expression of p62, alpha-SMA, and TGF-ß1, which validated that fluorofenidone could inhibit the development of paraquat-induced pulmonary fibrosis by increasing autophagy. CONCLUSIONS In conclusion, metabolomics combined with network pharmacology research strategy revealed that fluorofenidone has a multi-target and multi-path mechanism of action in the treatment of pulmonary fibrosis.


Subject(s)
Pulmonary Fibrosis/drug therapy , Pyridones/therapeutic use , Animals , Autophagy , Cadherins/metabolism , Disease Models, Animal , Gene Regulatory Networks , Humans , Lactosylceramides/metabolism , Male , Metabolomics , Paraquat , Pulmonary Fibrosis/chemically induced , Rats , Rats, Sprague-Dawley , Signal Transduction
6.
Int J Mol Sci ; 22(12)2021 Jun 13.
Article in English | MEDLINE | ID: mdl-34199317

ABSTRACT

Empagliflozin, an established treatment for type 2 diabetes (T2DM), has shown beneficial effects on liver steatosis and fibrosis in animals and in humans with T2DM, non-alcoholic fatty liver disease (NAFLD) and steatohepatitis (NASH). However, little is known about the effects of empagliflozin on liver function in advanced NASH with liver fibrosis and without diabetes. This study aimed to assess the effects of empagliflozin on hepatic and metabolic outcomes in a diet-induced obese (DIO) and insulin-resistant but non-diabetic biopsy-confirmed mouse model of advanced NASH. Male C57BL/6JRj mice with a biopsy-confirmed steatosis and fibrosis on AMLN diet (high fat, fructose and cholesterol) for 36-weeks were randomized to receive for 12 weeks: (a) Empagliflozin (10 mg/kg/d p.o.), or (b) vehicle. Metabolic outcomes, liver pathology, markers of Kupffer and stellate cell activation and lipidomics were assessed at the treatment completion. Empagliflozin did not affect the body weight, body composition or insulin sensitivity (assessed by intraperitoneal insulin tolerance test), but significantly improved glucose homeostasis as assessed by oral glucose tolerance test in DIO-NASH mice. Empagliflozin improved modestly the NAFLD activity score compared with the vehicle, mainly by improving inflammation and without affecting steatosis, the fibrosis stage and markers of Kupffer and stellate cell activation. Empagliflozin reduced the hepatic concentrations of pro-inflammatory lactosylceramides and increased the concentrations of anti-inflammatory polyunsaturated triglycerides. Empagliflozin exerts beneficial metabolic and hepatic (mainly anti-inflammatory) effects in non-diabetic DIO-NASH mice and thus may be effective against NASH even in non-diabetic conditions.


Subject(s)
Benzhydryl Compounds/therapeutic use , Glucosides/therapeutic use , Liver/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Antigens, CD/metabolism , Benzhydryl Compounds/pharmacology , Biopsy , Body Composition/drug effects , Body Weight/drug effects , Disease Models, Animal , Glucose/metabolism , Glucosides/pharmacology , Homeostasis/drug effects , Insulin Resistance , Lactosylceramides/metabolism , Lipidomics , Liver/drug effects , Liver/pathology , Mice, Inbred C57BL , Mice, Obese , Non-alcoholic Fatty Liver Disease/pathology , Triglycerides/metabolism
7.
Int J Mol Sci ; 22(4)2021 Feb 12.
Article in English | MEDLINE | ID: mdl-33673027

ABSTRACT

Lactosylceramide (LacCer), also known as CD17/CDw17, is a member of a large family of small molecular weight compounds known as glycosphingolipids. It plays a pivotal role in the biosynthesis of glycosphingolipids, primarily by way of serving as a precursor to the majority of its higher homolog sub-families such as gangliosides, sulfatides, fucosylated-glycosphingolipids and complex neutral glycosphingolipids-some of which confer "second-messenger" and receptor functions. LacCer is an integral component of the "lipid rafts," serving as a conduit to transduce external stimuli into multiple phenotypes, which may contribute to mortality and morbidity in man and in mouse models of human disease. LacCer is synthesized by the action of LacCer synthase (ß-1,4 galactosyltransferase), which transfers galactose from uridine diphosphate galactose (UDP-galactose) to glucosylceramide (GlcCer). The convergence of multiple physiologically relevant external stimuli/agonists-platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF), stress, cigarette smoke/nicotine, tumor necrosis factor-α (TNF-α), and in particular, oxidized low-density lipoprotein (ox-LDL)-on ß-1,4 galactosyltransferase results in its phosphorylation or activation, via a "turn-key" reaction, generating LacCer. This newly synthesized LacCer activates NADPH (nicotinamide adenine dihydrogen phosphate) oxidase to generate reactive oxygen species (ROS) and a highly "oxidative stress" environment, which trigger a cascade of signaling molecules and pathways and initiate diverse phenotypes like inflammation and atherosclerosis. For instance, LacCer activates an enzyme, cytosolic phospholipase A2 (cPLA2), which cleaves arachidonic acid from phosphatidylcholine. In turn, arachidonic acid serves as a precursor to eicosanoids and prostaglandin, which transduce a cascade of reactions leading to inflammation-a major phenotype underscoring the initiation and progression of several debilitating diseases such as atherosclerosis and cancer. Our aim here is to present an updated account of studies made in the field of LacCer metabolism and signaling using multiple animal models of human disease, human tissue, and cell-based studies. These advancements have led us to propose that previously unrelated phenotypes converge in a LacCer-centric manner. This LacCer synthase/LacCer-induced "oxidative stress" environment contributes to inflammation, atherosclerosis, skin conditions, hair greying, cardiovascular disease, and diabetes due to mitochondrial dysfunction. Thus, targeting LacCer synthase may well be the answer to remedy these pathologies.


Subject(s)
Antigens, CD/metabolism , Atherosclerosis/metabolism , Cardiovascular Diseases/metabolism , Diabetes Mellitus/metabolism , Lactosylceramides/metabolism , Oxidative Stress , Signal Transduction , Skin Diseases/metabolism , Animals , Antigens, CD/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/therapy , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Cardiovascular Diseases/therapy , Cytokines/genetics , Cytokines/metabolism , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Diabetes Mellitus/therapy , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation/therapy , Lactosylceramides/genetics , Mice , Skin Diseases/genetics , Skin Diseases/pathology , Skin Diseases/therapy
8.
Org Biomol Chem ; 18(19): 3724-3733, 2020 05 20.
Article in English | MEDLINE | ID: mdl-32364197

ABSTRACT

Glycosphingolipids (GSLs) are a group of molecules composed of a hydrophilic glycan part and a hydrophobic ceramide creating a diverse family. GSLs are de novo synthesised from ceramides at the endoplasmic reticulum and Golgi apparatus, and transported to the outer surface of the plasma membrane. It has been known that the glycan structures of GSLs change reflecting disease states. We envisioned that analysing the glycan pattern of GSLs enables distinguishing diseases. For this purpose, we utilised a fluorescently tagged compound, LacCerBODIPY (1). At first, compound 1 was taken up by cultured PC12D cells and transformed into various GSLs. As a result, changes in the GSL patterns of differentiation states of the cells were successfully observed by using an analysis platform, nano-liquid chromatography (LC)-fluorescence detection (FLD)-electrospray ionisation (ESI)-mass spectrometry (MS), which could quantify and provide molecular ions simultaneously. We found that compound 1 remained for about 10 min on the plasma membrane before it was converted into other GSLs. We therefore investigated a more rapid way to discriminate different cellular states by fluorescence recovery after photobleaching, which revealed that it is possible to distinguish the differentiation states as well.


Subject(s)
Boron Compounds/metabolism , Cell Membrane/metabolism , Lactosylceramides/metabolism , Polysaccharides/metabolism , Animals , Boron Compounds/chemistry , Cell Membrane/chemistry , Lactosylceramides/chemistry , Molecular Structure , PC12 Cells , Polysaccharides/chemistry , Rats
9.
Int J Mol Sci ; 20(23)2019 Nov 30.
Article in English | MEDLINE | ID: mdl-31801289

ABSTRACT

The development and progression of colorectal cancer (CRC), a major cause of cancer-related death in the western world, is accompanied with alterations of sphingolipid (SL) composition in colon tumors. A number of enzymes involved in the SL metabolism have been found to be deregulated in human colon tumors, in experimental rodent studies, and in human colon cancer cells in vitro. Therefore, the enzymatic pathways that modulate SL levels have received a significant attention, due to their possible contribution to CRC development, or as potential therapeutic targets. Many of these enzymes are associated with an increased sphingosine-1-phosphate/ceramide ratio, which is in turn linked with increased colon cancer cell survival, proliferation and cancer progression. Nevertheless, more attention should also be paid to the more complex SLs, including specific glycosphingolipids, such as lactosylceramides, which can be also deregulated during CRC development. In this review, we focus on the potential roles of individual SLs/SL metabolism enzymes in colon cancer, as well as on the pros and cons of employing the current in vitro models of colon cancer cells for lipidomic studies investigating the SL metabolism in CRC.


Subject(s)
Colonic Neoplasms/enzymology , Gene Expression Regulation, Neoplastic , Lactosylceramides/metabolism , Lipid Metabolism/genetics , Sphingolipids/metabolism , Acid Ceramidase/genetics , Acid Ceramidase/metabolism , Alkaline Ceramidase/genetics , Alkaline Ceramidase/metabolism , Animals , Ceramides/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Disease Models, Animal , Humans , Lysophospholipids/metabolism , Neutral Ceramidase/genetics , Neutral Ceramidase/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine N-Acyltransferase/genetics , Sphingosine N-Acyltransferase/metabolism , Tumor Cells, Cultured
10.
J Gen Virol ; 99(12): 1643-1657, 2018 12.
Article in English | MEDLINE | ID: mdl-30311874

ABSTRACT

Hepatitis C virus (HCV) infection is known to induce autophagy, but the mechanism of autophagy induced by HCV remains controversial. Here, we investigated the characteristics of autophagy induced by HCV infection. First, to examine the involvement of autophagy-related gene (ATG) proteins in HCV-induced LC3 lipidation, we established ATG5, ATG13 or ATG14 knockout (KO) Huh7.5.1 cell lines and confirmed that the accumulation of lipidated LC3 was induced in an ATG13- and ATG14-independent manner. On the other hand, HCV infectivity was not influenced by deficiencies in these genes. We also confirmed that LC3-positive dots were co-localized with ubiquitinated aggregates, and deficiency of ATG5 or ATG14 enhanced the accumulation of ubiquitinated aggregates compared to that in the restored cells, suggesting that HCV infection induces ATG5- and ATG14-dependent selective autophagy. Moreover, LC3-positive ubiquitinated aggregates accumulated near the site of the replication complex. We further examined autophagy flux in cells replicating HCV RNA using bafilomycin or E64d, and found that the increase of LC3 lipidation by treatment with bafilomycin or E64d was impaired in HCV-replicating cells, suggesting that autophagy flux is inhibited by the progress of HCV infection. Our present study suggests that (1) HCV RNA replication induces selective autophagy and (2) the progress of HCV infection impairs autophagy flux.


Subject(s)
Autophagy , Hepacivirus/growth & development , Hepacivirus/immunology , Hepatocytes/virology , Virus Replication , Autophagy-Related Proteins/metabolism , Cell Line , Humans , Lactosylceramides/metabolism
11.
J Proteome Res ; 16(8): 2802-2810, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28628327

ABSTRACT

Niemann-Pick disease type C (NPC) is an autosomal recessive lipid storage disorder, and the majority of cases are caused by mutations in the NPC1 gene. In this study, we clarified how a single gene mutation in the NPC1 gene impacts the cellular glycome by analyzing the total glycomic expression profile of Chinese hamster ovary cell mutants defective in the Npc1 gene (Npc1 KO CHO cells). A number of glycomic alterations were identified, including increased expression of lactosylceramide, GM1, GM2, GD1, various neolacto-series glycosphingolipids, and sialyl-T (O-glycan), which was found to be the major sialylated protein-bound glycan, as well as various N-glycans, which were commonly both fucosylated and sialylated. We also observed significant increases in the total amounts of free oligosaccharides (fOSs), especially in the unique complex- and hybrid-type fOSs. Treatment of Npc1 KO CHO cells with 2-hydroxypropyl-ß-cyclodextrin (HPBCD), which can reduce cholesterol and glycosphingolipid (GSL) storage, did not affect the glycomic alterations observed in the GSL-, N-, and O-glycans of Npc1 KO CHO cells. However, HPBCD treatment corrected the glycomic alterations observed in fOSs to levels observed in wild-type cells.


Subject(s)
Glycomics , Mutation , Niemann-Pick Disease, Type C/genetics , Animals , Antigens, CD/metabolism , CHO Cells , Cricetulus , Glycosphingolipids/metabolism , Lactosylceramides/metabolism , Membrane Glycoproteins/analysis , Membrane Glycoproteins/genetics , Polysaccharides/analysis , beta-Cyclodextrins/pharmacology
12.
J Biol Chem ; 291(37): 19245-56, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27435676

ABSTRACT

Endorepellin, the C-terminal domain of perlecan, is an angiostatic molecule that acts as a potent inducer of autophagy via its interaction with VEGFR2. In this study, we examined the effect of endorepellin on endothelial cells using atomic force microscopy. Soluble endorepellin caused morphological and biophysical changes such as an increase in cell surface roughness and cell height. Surprisingly, these changes were not accompanied by alterations in the endothelial cell elastic modulus. We discovered that endorepellin-induced autophagic flux led to co-localization of mammalian target of rapamycin with LC3-positive autophagosomes. Endorepellin functioned upstream of AMP-activated kinase α, as compound C, an inhibitor of AMP-activated kinase α, abrogated endorepellin-mediated activation and co-localization of Beclin 1 and LC3, thereby reducing autophagic progression. Functionally, we discovered that both endorepellin and Torin 1, a canonical autophagic inducer, blunted ex vivo angiogenesis. We conclude that autophagy is a novel mechanism by which endorepellin promotes angiostasis independent of nutrient deprivation.


Subject(s)
Autophagy , Heparan Sulfate Proteoglycans/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Peptide Fragments/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Adenylate Kinase/metabolism , Beclin-1/metabolism , Elastic Modulus , Human Umbilical Vein Endothelial Cells/ultrastructure , Humans , Lactosylceramides/metabolism
13.
J Immunol ; 195(12): 5551-60, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26538397

ABSTRACT

The ETS factor Friend leukemia virus integration 1 (FLI1) is a key modulator of lupus disease expression. Overexpressing FLI1 in healthy mice results in the development of an autoimmune kidney disease similar to that observed in lupus. Lowering the global levels of FLI1 in two lupus strains (Fli1(+/-)) significantly improved kidney disease and prolonged survival. T cells from MRL/lpr Fli1(+/-) lupus mice have reduced activation and IL-4 production, neuraminidase 1 expression, and the levels of the glycosphingolipid lactosylceramide. In this study, we demonstrate that MRL/lpr Fli1(+/-) mice have significantly decreased renal neuraminidase 1 and lactosylceramide levels. This corresponds with a significant decrease in the number of total CD3(+) cells, as well as CD4(+) and CD44(+)CD62L(-) T cell subsets in the kidney of MRL/lpr Fli1(+/-) mice compared with the Fli1(+/+) nephritic mice. We further demonstrate that the percentage of CXCR3(+) T cells and Cxcr3 message levels in T cells are significantly decreased and correspond with a decrease in renal CXCR3(+) cells and in Cxcl9 and Cxcl10 expression in the MRL/lpr Fli1(+/-) compared with the Fli1(+/+) nephritic mice. Our results suggest that reducing the levels of FLI1 in MRL/lpr mice may be protective against development of nephritis in part through downregulation of CXCR3, reducing renal T cell infiltration and glycosphingolipid levels.


Subject(s)
Glycosphingolipids/metabolism , Kidney/physiology , Nephritis/drug therapy , Proto-Oncogene Protein c-fli-1/metabolism , Receptors, CXCR3/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD/metabolism , Cell Movement/drug effects , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Chemokine CXCL9/genetics , Chemokine CXCL9/metabolism , Gene Expression Regulation , Humans , Kidney/drug effects , Lactosylceramides/metabolism , Mice , Mice, Inbred MRL lpr , Mice, Knockout , Nephritis/immunology , Neuraminidase/metabolism , Proto-Oncogene Protein c-fli-1/genetics , Receptors, CXCR3/genetics
14.
J Lipid Res ; 57(4): 546-62, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26900161

ABSTRACT

Sphingolipids have been implicated as key mediators of cell-stress responses and effectors of mitochondrial function. To investigate potential mechanisms underlying mitochondrial dysfunction, an important contributor to diabetic cardiomyopathy, we examined alterations of cardiac sphingolipid metabolism in a mouse with streptozotocin-induced type 1 diabetes. Diabetes increased expression of desaturase 1, (dihydro)ceramide synthase (CerS)2, serine palmitoyl transferase 1, and the rate of ceramide formation by mitochondria-resident CerSs, indicating an activation of ceramide biosynthesis. However, the lack of an increase in mitochondrial ceramide suggests concomitant upregulation of ceramide-metabolizing pathways. Elevated levels of lactosylceramide, one of the initial products in the formation of glycosphingolipids were accompanied with decreased respiration and calcium retention capacity (CRC) in mitochondria from diabetic heart tissue. In baseline mitochondria, lactosylceramide potently suppressed state 3 respiration and decreased CRC, suggesting lactosylceramide as the primary sphingolipid responsible for mitochondrial defects in diabetic hearts. Moreover, knocking down the neutral ceramidase (NCDase) resulted in an increase in lactosylceramide level, suggesting a crosstalk between glucosylceramide synthase- and NCDase-mediated ceramide utilization pathways. These data suggest the glycosphingolipid pathway of ceramide metabolism as a promising target to correct mitochondrial abnormalities associated with type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Lactosylceramides/metabolism , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Animals , Cell Respiration , Diabetes Mellitus, Type 1/enzymology , Diabetes Mellitus, Type 1/physiopathology , Gene Expression Regulation, Enzymologic , Gene Knockdown Techniques , Heart/physiopathology , Hydrolysis , Male , Mice , Mice, Inbred C57BL , Neutral Ceramidase/deficiency , Neutral Ceramidase/genetics , Neutral Ceramidase/metabolism
15.
Biochim Biophys Acta ; 1851(1): 90-7, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24968752

ABSTRACT

Populations of glycolipids change markedly during leukocyte differentiation, suggesting that these molecules are involved in biological functions. About 70% of the glycosphingolipids in human neutrophils are lactosylceramide, a molecule also expressed on monocytes and dendritic cells, but not on lymphocytes. In contrast, phosphatidylglucoside is mainly expressed on neutrophils. STED microscopic analysis showed that phosphatidylglucoside and lactosylceramide form different domains on plasma membranes of neutrophils, with phosphatidylglucoside preferentially expressed along the neutrophil differentiation pathway. Phosphatidylglucoside was found to mediate the differentiation of HL-60 cells into the neutrophilic lineage, and to be involved in FAS-dependent neutrophil apoptosis. In contrast, lactosylceramide was only expressed on mature neutrophils. Complexes of lactosylceramide and the Src family kinase Lyn form membrane microdomains. LacCer-enriched membrane microdomains mediate neutrophil innate immune responses; e.g. chemotaxis, phagocytosis, and superoxide generation. C24 fatty acid chains of LacCer are indispensable for the formation of LacCer-Lyn complexes and for LacCer-dependent functions. Moreover, Lyn-coupled LacCer-enriched microdomains serve as signal transduction platforms for αMß2 integrin-mediated phagocytosis. This review describes the organization and potential functions of glycolipids in phagocytes, as well as the roles of both phosphatidylglucoside and lactosylceramide in neutrophils. This article is part of a Special Issue entitled Linking transcription to physiology in lipidomics.


Subject(s)
Glycolipids/metabolism , Membrane Microdomains/metabolism , Phagocytes/metabolism , Antigens, CD/metabolism , Cell Differentiation/physiology , Glycerophospholipids/metabolism , Humans , Lactosylceramides/metabolism , Neutrophils/metabolism
16.
Hum Mol Genet ; 23(4): 843-54, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24064337

ABSTRACT

Gaucher disease has recently received wide attention due to the unexpected discovery that it is a genetic risk factor for Parkinson's disease. Gaucher disease is caused by the defective activity of the lysosomal enzyme, glucocerebrosidase (GCase; GBA1), resulting in intracellular accumulation of the glycosphingolipids, glucosylceramide and psychosine. The rare neuronopathic forms of GD (nGD) are characterized by profound neurological impairment and neuronal cell death. We have previously described the progression of neuropathological changes in a mouse model of nGD. We now examine the relationship between glycosphingolipid accumulation and initiation of pathology at two pre-symptomatic stages of the disease in four different brain areas which display differential degrees of susceptibility to GCase deficiency. Liquid chromatography electrospray ionization tandem mass spectrometry demonstrated glucosylceramide and psychosine accumulation in nGD brains prior to the appearance of neuroinflammation, although only glucosylceramide accumulation correlated with neuroinflammation and neuron loss. Levels of other sphingolipids, including the pro-apoptotic lipid, ceramide, were mostly unaltered. Transmission electron microscopy revealed that glucosylceramide accumulation occurs in neurons, mostly in the form of membrane-delimited pseudo-tubules located near the nucleus. Highly disrupted glucosylceramide-storing cells, which are likely degenerating neurons containing massive inclusions, numerous autophagosomes and unique ultrastructural features, were also observed. Together, our results indicate that a certain level of neuronal glucosylceramide storage is required to trigger neuropathological changes in affected brain areas, while other brain areas containing similar glucosylceramide levels are unaltered, presumably because of intrinsic differences in neuronal properties, or in the neuronal environment, between various brain regions.


Subject(s)
Gaucher Disease/metabolism , Glucosylceramides/metabolism , Nerve Degeneration/metabolism , Neurons/metabolism , Animals , Brain/metabolism , Brain/pathology , Gaucher Disease/pathology , Glucosylceramidase/deficiency , Glucosylceramidase/genetics , Humans , Lactosylceramides/metabolism , Mice , Mice, Knockout , Neurons/pathology , Psychosine/metabolism , Sphingomyelins/metabolism
17.
J Neurosci Res ; 94(11): 1246-60, 2016 11.
Article in English | MEDLINE | ID: mdl-27638607

ABSTRACT

Globoid cell leukodystrophy (GLD) is a rare, rapidly progressing childhood leukodystrophy triggered by deficit of the lysosomal enzyme galactosylceramidase (GALC) and characterized by the accumulation of galactosylsphingosine (psychosine; PSY) in the nervous system. PSY is a cytotoxic sphingolipid, which leads to widespread degeneration of oligodendrocytes and Schwann cells, causing demyelination. Here we report on autophagy in the human oligodendrocyte cell line MO3.13 treated with PSY and exploitation of Li as an autophagy modulator to rescue cell viability. We demonstrate that PSY causes upregulation of the autophagic flux at the level of autophagosome and autolysosome formation and LC3-II expression. We show that pretreatment with Li, a drug clinically used to treat bipolar disorders, can further stimulate autophagy, improving cell tolerance to PSY. This Li protective effect is found not to be linked to reduction of PSY-induced oxidative stress and might not stem from a reduction of PSY accumulation. These data provide novel information on the intracellular pathways activated during PSY-induced toxicity and suggest the autophagy pathway as a promising novel therapeutic target for ameliorating the GLD phenotype. © 2016 Wiley Periodicals, Inc.


Subject(s)
Autophagy/drug effects , Lithium/pharmacology , Oligodendroglia/drug effects , Psychosine/pharmacology , Analysis of Variance , Annexin A5/metabolism , Cell Line, Transformed , Cell Survival/drug effects , Dose-Response Relationship, Drug , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lactosylceramides/genetics , Lactosylceramides/metabolism , Psychosine/metabolism , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein/metabolism , Signal Transduction/drug effects , Transfection , Tubulin/genetics , Tubulin/metabolism
18.
J Lipid Res ; 56(1): 129-41, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25418321

ABSTRACT

Lactosylceramide [LacCer; ß-Gal-(1-4)-ß-Glc-(1-1)-Cer] has been shown to contain very long fatty acids that specifically modulate neutrophil properties. The interactions between LacCer and proteins and their role in cell signaling processes were assessed by synthesizing two molecular species of azide-photoactivable tritium-labeled LacCer having acyl chains of different lengths. The lengths of the two acyl chains corresponded to those of a short/medium and very long fatty acid, comparable to the lengths of stearic and lignoceric acids, respectively. These derivatives, designated C18-[(3)H]LacCer-(N3) and C24-[(3)H]LacCer-(N3), were incorporated into the lipid rafts of plasma membranes of neutrophilic differentiated HL-60 (D-HL-60) cells. C24-[(3)H]LacCer-(N3), but not C18-[(3)H]LacCer-(N3), induced the phosphorylation of Lyn and promoted phagocytosis. Incorporation of C24-[(3)H]LacCer-(N3) into plasma membranes, followed by illumination, resulted in the formation of several tritium-labeled LacCer-protein complexes, including the LacCer-Lyn complex, into plasma membrane lipid rafts. Administration of C18-[(3)H]LacCer-(N3) to cells, however, did not result in the formation of the LacCer-Lyn complex. These results suggest that LacCer derivatives mimic the biological properties of natural LacCer species and can be utilized as tools to study LacCer-protein interactions, and confirm a specific direct interaction between LacCer species containing very long fatty acids, and Lyn protein, associated with the cytoplasmic layer via myristic/palmitic chains.


Subject(s)
Antigens, CD/metabolism , Lactosylceramides/metabolism , Membrane Microdomains/metabolism , Neutrophils/cytology , Signal Transduction , src-Family Kinases/metabolism , Animals , Antigens, CD/chemistry , Antigens, CD/pharmacology , Azides/chemistry , Cell Survival/drug effects , HL-60 Cells , Humans , Lactosylceramides/chemistry , Lactosylceramides/pharmacology , Membrane Microdomains/drug effects , Neutrophils/immunology , Phagocytosis/drug effects , Phosphorylation/drug effects , Protein Binding , Signal Transduction/drug effects
19.
J Biol Chem ; 289(30): 20845-57, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24928515

ABSTRACT

Sphingosine kinase 2 (SPK2) and autophagy are both involved in brain preconditioning, but whether preconditioning-induced SPK2 up-regulation and autophagy activation are linked mechanistically remains to be elucidated. In this study, we used in vitro and in vivo models to explore the role of SPK2-mediated autophagy in isoflurane and hypoxic preconditioning. In primary mouse cortical neurons, both isoflurane and hypoxic preconditioning induced autophagy. Isoflurane and hypoxic preconditioning protected against subsequent oxygen glucose deprivation or glutamate injury, whereas pretreatment with autophagy inhibitors (3-methyladenine or KU55933) abolished preconditioning-induced tolerance. Pretreatment with SPK2 inhibitors (ABC294640 and SKI-II) or SPK2 knockdown prevented preconditioning-induced autophagy. Isoflurane also induced autophagy in mouse in vivo as shown by Western blots for LC3 and p62, LC3 immunostaining, and electron microscopy. Isoflurane-induced autophagy in mice lacking the SPK1 isoform (SPK1(-/-)), but not in SPK2(-/-)mice. Sphingosine 1-phosphate and the sphingosine 1-phosphate receptor agonist FTY720 did not protect against oxygen glucose deprivation in cultured neurons and did not alter the expression of LC3 and p62, suggesting that SPK2-mediated autophagy and protections are not S1P-dependent. Beclin 1 knockdown abolished preconditioning-induced autophagy, and SPK2 inhibitors abolished isoflurane-induced disruption of the Beclin 1/Bcl-2 association. These results strongly indicate that autophagy is involved in isoflurane preconditioning both in vivo and in vitro and that SPK2 contributes to preconditioning-induced autophagy, possibly by disrupting the Beclin 1/Bcl-2 interaction.


Subject(s)
Autophagy , Cerebral Cortex/metabolism , Ischemic Preconditioning , Neurons/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Adamantane/analogs & derivatives , Adamantane/pharmacology , Anesthetics, Inhalation/pharmacology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Cells, Cultured , Cerebral Cortex/cytology , Enzyme Inhibitors/pharmacology , Isoflurane/pharmacology , Lactosylceramides/genetics , Lactosylceramides/metabolism , Mice , Mice, Knockout , Morpholines/pharmacology , Neurons/cytology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyridines/pharmacology , Pyrones/pharmacology , Transcription Factor TFIIH , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Circulation ; 129(23): 2403-13, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24710030

ABSTRACT

BACKGROUND: Glycosphingolipids, integral components of the cell membrane, have been shown to serve as messengers, transducing growth factor-initiated phenotypes. Here, we have examined whether inhibition of glycosphingolipid synthesis could ameliorate atherosclerosis and arterial stiffness in transgenic mice and rabbits. METHODS AND RESULTS: Apolipoprotein E(-/-) mice (12 weeks of age; n=6) were fed regular chow or a Western diet (1.25% cholesterol, 2% fat). Mice were fed 5 or 10 mg/kg of an inhibitor of glycosphingolipid synthesis, D-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (D-PDMP), solubilized in vehicle (5% Tween-80 in PBS); the placebo group received vehicle only. At 20 and 36 weeks of age, serial echocardiography was performed to measure aortic intima-media thickening. Aortic pulse-wave velocity measured vascular stiffness. Feeding mice a Western diet markedly increased aortic pulse-wave velocity, intima-media thickening, oxidized low-density lipoprotein, Ca(2+) deposits, and glucosylceramide and lactosylceramide synthase activity. These were dose-dependently decreased by feeding D-PDMP. In liver, D-PDMP decreased cholesterol and triglyceride levels by raising the expression of SREBP2, low-density lipoprotein receptor, HMGCo-A reductase, and the cholesterol efflux genes (eg, ABCG5, ABCG8). D-PDMP affected very-low-density lipoprotein catabolism by increasing the gene expression for lipoprotein lipase and very-low-density lipoprotein receptor. Rabbits fed a Western diet for 90 days had extensive atherosclerosis accompanied by a 17.5-fold increase in total cholesterol levels and a 3-fold increase in lactosylceramide levels. This was completely prevented by feeding D-PDMP. CONCLUSIONS: Inhibition of glycosphingolipid synthesis ameliorates atherosclerosis and arterial stiffness in apolipoprotein E(-/-) mice and rabbits. Thus, inhibition of glycosphingolipid synthesis may be a novel approach to ameliorate atherosclerosis and arterial stiffness.


Subject(s)
Coronary Artery Disease/drug therapy , Coronary Artery Disease/metabolism , Diet, High-Fat , Glycosphingolipids/biosynthesis , Morpholines/pharmacology , Vascular Stiffness/drug effects , Animals , Antigens, CD/metabolism , Aorta/diagnostic imaging , Aorta/drug effects , Aorta/metabolism , Apolipoproteins E/genetics , Calcium/metabolism , Cholesterol, Dietary/pharmacology , Coronary Artery Disease/genetics , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Glucosylceramides/metabolism , Glycosphingolipids/metabolism , Hyperlipidemias/drug therapy , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Lactosylceramides/metabolism , Lipoproteins, LDL/metabolism , Male , Mice , Mice, Knockout , Models, Cardiovascular , Pulsatile Flow/drug effects , Rabbits , Ultrasonography
SELECTION OF CITATIONS
SEARCH DETAIL