Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(22)2021 Nov 22.
Article in English | MEDLINE | ID: mdl-34830477

ABSTRACT

Singapore grouper iridovirus (SGIV), belonging to genus Ranavirus, family Iridoviridae, causes great economic losses in the aquaculture industry. Previous studies demonstrated the lipid composition of intracellular unenveloped viruses, but the changes in host-cell glyceophospholipids components and the roles of key enzymes during SGIV infection still remain largely unknown. Here, the whole cell lipidomic profiling during SGIV infection was analyzed using UPLC-Q-TOF-MS/MS. The lipidomic data showed that glycerophospholipids (GPs), including phosphatidylcholine (PC), phosphatidylserine (PS), glycerophosphoinositols (PI) and fatty acids (FAs) were significantly elevated in SGIV-infected cells, indicating that SGIV infection disturbed GPs homeostasis, and then affected the metabolism of FAs, especially arachidonic acid (AA). The roles of key enzymes, such as cytosolic phospholipase A2 (cPLA2), 5-Lipoxygenase (5-LOX), and cyclooxygenase (COX) in SGIV infection were further investigated using the corresponding specific inhibitors. The inhibition of cPLA2 by AACOCF3 decreased SGIV replication, suggesting that cPLA2 might play important roles in the process of SGIV infection. Consistent with this result, the ectopic expression of EccPLA2α or knockdown significantly enhanced or suppressed viral replication in vitro, respectively. In addition, the inhibition of both 5-LOX and COX significantly suppressed SGIV replication, indicating that AA metabolism was essential for SGIV infection. Taken together, our results demonstrated for the first time that SGIV infection in vitro disturbed GPs homeostasis and cPLA2 exerted crucial roles in SGIV replication.


Subject(s)
Fishes/virology , Iridovirus/genetics , Phospholipases A2, Cytosolic/genetics , Virus Replication/genetics , Animals , Aquaculture , Arachidonate 5-Lipoxygenase/genetics , Fishes/genetics , Glycerophospholipids/genetics , Iridovirus/pathogenicity , Phosphatidylcholines/genetics , Phosphatidylserines/genetics , Singapore
2.
J Biol Chem ; 297(1): 100851, 2021 07.
Article in English | MEDLINE | ID: mdl-34089703

ABSTRACT

Phospholipase A1 (PLA1) hydrolyzes the fatty acids of glycerophospholipids, which are structural components of the cellular membrane. Genetic mutations in DDHD1, an intracellular PLA1, result in hereditary spastic paraplegia (HSP) in humans. However, the regulation of DDHD1 activity has not yet been elucidated in detail. In the present study, we examined the phosphorylation of DDHD1 and identified the responsible protein kinases. We performed MALDI-TOF MS/MS analysis and Phos-tag SDS-PAGE in alanine-substitution mutants in HEK293 cells and revealed multiple phosphorylation sites in human DDHD1, primarily Ser8, Ser11, Ser723, and Ser727. The treatment of cells with a protein phosphatase inhibitor induced the hyperphosphorylation of DDHD1, suggesting that multisite phosphorylation occurred not only at these major, but also at minor sites. Site-specific kinase-substrate prediction algorithms and in vitro kinase analyses indicated that cyclin-dependent kinase CDK1/cyclin A2 phosphorylated Ser8, Ser11, and Ser727 in DDHD1 with a preference for Ser11 and that CDK5/p35 also phosphorylated Ser11 and Ser727 with a preference for Ser11. In addition, casein kinase CK2α1 was found to phosphorylate Ser104, although this was not a major phosphorylation site in cultivated HEK293 cells. The evaluation of the effects of phosphorylation revealed that the phosphorylation mimic mutants S11/727E exhibit only 20% reduction in PLA1 activity. However, the phosphorylation mimics were mainly localized to focal adhesions, whereas the phosphorylation-resistant mutants S11/727A were not. This suggested that phosphorylation alters the subcellular localization of DDHD1 without greatly affecting its PLA1 activity.


Subject(s)
CDC2 Protein Kinase/genetics , Cyclin A2/genetics , Phospholipases A1/genetics , CDC2 Protein Kinase/chemistry , Cell Membrane/chemistry , Cell Membrane/genetics , Cyclin A2/chemistry , Glycerophospholipids/chemistry , Glycerophospholipids/genetics , HEK293 Cells , Humans , Phospholipases A1/chemistry , Phospholipases A1/metabolism , Phosphorylation/genetics , Spastic Paraplegia, Hereditary/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
3.
Biomolecules ; 10(10)2020 09 29.
Article in English | MEDLINE | ID: mdl-33003344

ABSTRACT

Phosphatidic acid (PA) is a glycerophospholipid intermediate in the triglyceride synthesis pathway that has incredibly important structural functions as a component of cell membranes and dynamic effects on intracellular and intercellular signaling pathways. Although there are many pathways to synthesize and degrade PA, a family of PA phosphohydrolases (lipin family proteins) that generate diacylglycerol constitute the primary pathway for PA incorporation into triglycerides. Previously, it was believed that the pool of PA used to synthesize triglyceride was distinct, compartmentalized, and did not widely intersect with signaling pathways. However, we now know that modulating the activity of lipin 1 has profound effects on signaling in a variety of cell types. Indeed, in most tissues except adipose tissue, lipin-mediated PA phosphohydrolase activity is far from limiting for normal rates of triglyceride synthesis, but rather impacts critical signaling cascades that control cellular homeostasis. In this review, we will discuss how lipin-mediated control of PA concentrations regulates metabolism and signaling in mammalian organisms.


Subject(s)
Glycerophospholipids/genetics , Phosphatidate Phosphatase/genetics , Triglycerides/biosynthesis , Adipose Tissue/metabolism , Glycerophospholipids/metabolism , Humans , Metabolic Networks and Pathways/genetics , Muscle, Skeletal/metabolism , Phosphatidate Phosphatase/ultrastructure , Phosphatidic Acids/biosynthesis , Phosphatidic Acids/metabolism , Protein Conformation , Triglycerides/metabolism
4.
Biomolecules ; 10(10)2020 10 19.
Article in English | MEDLINE | ID: mdl-33086624

ABSTRACT

The phospholipase A2 (PLA2) superfamily contains more than 50 enzymes in mammals that are subdivided into several distinct families on a structural and biochemical basis. In principle, PLA2 has the capacity to hydrolyze the sn-2 position of glycerophospholipids to release fatty acids and lysophospholipids, yet several enzymes in this superfamily catalyze other reactions rather than or in addition to the PLA2 reaction. PLA2 enzymes play crucial roles in not only the production of lipid mediators, but also membrane remodeling, bioenergetics, and body surface barrier, thereby participating in a number of biological events. Accordingly, disturbance of PLA2-regulated lipid metabolism is often associated with various diseases. This review updates the current state of understanding of the classification, enzymatic properties, and biological functions of various enzymes belonging to the PLA2 superfamily, focusing particularly on the novel roles of PLA2s in vivo.


Subject(s)
Cell Membrane/enzymology , Glycerophospholipids/genetics , Lipid Metabolism/genetics , Phospholipases A2/genetics , Animals , Cell Membrane/genetics , Fatty Acids/genetics , Fatty Acids/metabolism , Glycerophospholipids/metabolism , Humans , Lysophospholipids/metabolism , Mammals/genetics
5.
JCI Insight ; 52019 07 25.
Article in English | MEDLINE | ID: mdl-31343987

ABSTRACT

BACKGROUND: Acute graft-versus-host disease (aGvHD) is a major factor that limits the successful outcomes of allogeneic hematopoietic cell transplantation (alloHSCT). Currently there are few validated biomarkers that can help predict the risk of aGvHD in clinical settings. METHODS: We performed an integrated metabolomics and transcriptomics study and identified biomarkers that distinguish alloHSCT recipients with aGvHD from alloHSCT recipients without aGvHD in two separate cohorts. RESULTS: Pathway analysis of 38 significantly altered metabolites and 1148 differentially expressed genes uncovered a distinctly altered glycerophospholipid (GPL) metabolism network. Subsequently, we developed an aGvHD risk score (GRS) based on 5 metabolites markers from GPL metabolism to predict the risk of aGvHD. GRS showed a positive predictive value of 92.2% and 89.6% in the training and validation cohorts, respectively. In addition, high GRS was correlated with poor overall survival. Gene expressions of GPL-related lipases were significantly altered in aGvHD samples, leading to dysregulated GPLs. CONCLUSIONS: Using integrative "Omic" analysis, we unraveled a comprehensive view of the molecular perturbations underlying the pathogenesis of aGvHD. Our work represents an initial investigation of a unique metabolic and transcriptomic network that may help identify aGvHD at an early stage and facilitate preemptive therapy. FUNDING: National Natural Science Foundation of China (NSFC; 81530047, 81870143, 81470321, 81770160, 81270567, 81270638, 81573396, 81703674). Shanghai Sailing Program from Science and Technology Commission Shanghai Municipality (17YF1424700). Scholarship from Shanghai Municipal Health and Family Planning Commission (2017BR012). Special Clinical Research in Health Industry in Shanghai (20184Y0054).


Subject(s)
Biomarkers , Glycerophospholipids/metabolism , Graft vs Host Disease/metabolism , Hematopoietic Stem Cell Transplantation/adverse effects , Metabolomics , Adolescent , Adult , Animals , China , Disease Models, Animal , Female , Glycerophospholipids/genetics , Humans , Male , Middle Aged , Transcriptome , Young Adult
6.
J Oleo Sci ; 68(7): 637-647, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31178465

ABSTRACT

In the present work, a high-performance liquid chromatographic method coupled with mass spectrometry (HILIC-HPLC /ESI-MS) was used for the characterization and the quantification of glycerophospholipids (GPLs) classes and their molecular species in three genetically different Tunisian apricot cultivars (bitter, sweet and semi-sweet apricots). The application of the proposed method to the analysis of apricot oil allowed to separate and identify 74 molecular species of GPLs. Phosphatidylcholine (PC) class was found to be the most abundant GLPs in the three seed oils (38.6-62.4%) especially in bitter apricot, followed by phosphatidylinositol (PI) and phosphatidylethanolamine (PE) classes with values of 8.3-38.9% and 1.7-25.4% respectively. Phosphatidic acid (PA), phosphatidylglycerol (PG) and lysophosphatidylcholine (LPC) compounds were minor ones with maximums of 11.3%, 9.8% and 9.2% respectively. The results we obtained for the three Tunisian apricot seed varieties clearly indicate that the phospholipids of Tunisian apricot are of great interest. In fact, the high content of phosphatidylcholine (PC) determines it as a suitable and valuable source for obtaining corresponding phospholipids concentrates.


Subject(s)
Glycerophospholipids/genetics , Glycerophospholipids/isolation & purification , Prunus armeniaca/chemistry , Prunus armeniaca/genetics , Seeds/chemistry , Chromatography, High Pressure Liquid/methods , Glycerophospholipids/analysis , Spectrometry, Mass, Electrospray Ionization/methods
7.
Stem Cells ; 37(4): 489-503, 2019 04.
Article in English | MEDLINE | ID: mdl-30599084

ABSTRACT

Iron homeostasis is crucial for a variety of biological processes, but the biological role of iron homeostasis in pluripotent stem cells (PSCs) remains largely unknown. The present study aimed to determine whether iron homeostasis is involved in maintaining the pluripotency of human PSCs (hPSCs). We found that the intracellular depletion of iron leads to a rapid downregulation of NANOG and a dramatic decrease in the self-renewal of hPSCs as well as spontaneous and nonspecific differentiation. Moreover, long-term depletion of iron can result in the remarkable cell death of hPSCs via apoptosis and necrosis pathways. Additionally, we found that the depletion of iron increased the activity of lipoprotein-associated phospholipase A2 (LP-PLA2) and the production of lysophosphatidylcholine, thereby suppressing NANOG expression by enhancer of zeste homolog 2-mediated trimethylation of histone H3 lysine 27. Consistently, LP-PLA2 inhibition abrogated iron depletion-induced loss of pluripotency and differentiation. Altogether, the findings of our study demonstrates that iron homeostasis, acting through glycerophospholipid metabolic pathway, is essential for the pluripotency and survival of hPSCs. Stem Cells 2019;37:489-503.


Subject(s)
Epigenesis, Genetic/genetics , Glycerophospholipids/genetics , Glycerophospholipids/metabolism , Iron/metabolism , Pluripotent Stem Cells/metabolism , Cell Differentiation , Homeostasis , Humans , Transfection
8.
Biomolecules ; 8(4)2018 11 22.
Article in English | MEDLINE | ID: mdl-30469542

ABSTRACT

In humans, the age of fibre cells differs across the ocular lens, ranging from those formed before birth in the core of the lens to those formed just prior to death in the outer cortex. The distribution of glycerophospholipids in the adult human lens should reflect this range; however, limited data currently exists to confirm this hypothesis. Accordingly, this study aimed to determine the distribution of glycerophospholipids in adult human lens using mass spectrometry imaging. To achieve this, 20-µm thick slices of two human lenses, aged 51 and 67 were analysed by matrix-assisted laser desorption ionisation imaging mass spectrometry. The data clearly indicate that intact glycerophospholipids such as phosphatidylethanolamine, phosphatidylserine, and phosphatidic acid are mainly present in the outer cortex region, corresponding to the youngest fibre cells, while lyso-phosphatidylethanolamine, likely produced by the degradation of phosphatidylethanolamine, is present in the nucleus (older fibre cells). This study adds further evidence to the relationship between fibre cell age and glycerophospholipid composition.


Subject(s)
Glycerophospholipids/metabolism , Lens, Crystalline/metabolism , Adult , Aged , Cell Lineage/genetics , Glycerophospholipids/genetics , Glycerophospholipids/isolation & purification , Humans , Lens, Crystalline/diagnostic imaging , Lysophospholipids/chemistry , Lysophospholipids/isolation & purification , Lysophospholipids/metabolism , Middle Aged , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
9.
FEBS Lett ; 592(8): 1330-1345, 2018 04.
Article in English | MEDLINE | ID: mdl-29265372

ABSTRACT

In most eukaryotes, including Saccharomyces cerevisiae, glycerophospholipids are the main membrane lipid constituents. Besides serving as general membrane 'building blocks', glycerophospholipids play an important role in determining the physical properties of the membrane, which are crucial for proper membrane function. To ensure optimal physical properties, membrane glycerophospholipid composition and synthesis are tightly regulated. This review will summarize our current knowledge of factors and processes determining the membrane glycerophospholipid composition of the reference eukaryote S. cerevisiae at the level of molecular species. Extrapolating from relevant model membrane data, we also discuss how modulation of the molecular species composition can regulate membrane physical properties.


Subject(s)
Cell Membrane/metabolism , Glycerophospholipids/biosynthesis , Models, Biological , Saccharomyces cerevisiae/metabolism , Cell Membrane/genetics , Glycerophospholipids/genetics , Saccharomyces cerevisiae/genetics
10.
J Bacteriol ; 199(13)2017 07 01.
Article in English | MEDLINE | ID: mdl-28439041

ABSTRACT

In this issue of the Journal of Bacteriology, V. W. Rowlett et al. unveil new Escherichia coli circuitry linking membrane glycerophospholipid (GPL) homeostasis to bacterial stress response and adaptation mechanisms (J Bacteriol 199:e00849-16, 2017, https://doi.org/10.1128/JB.00849-16). Glycerophospholipids comprise critical components of the dual-membrane envelope of Gram-negative bacteria and participate in many processes. The new evidence suggests that, in some instances, distinct E. coli GPL molecules function for distinct biochemistry and bacteria sense perturbations in membrane GPL concentrations to coordinate survival strategies. Understanding GPL sensing and remodeling mechanisms will be important moving forward, given the breadth of function for these molecules in bacteriology.


Subject(s)
Cell Membrane/physiology , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial/physiology , Glycerophospholipids/metabolism , Escherichia coli/genetics , Glycerophospholipids/genetics , Stress, Physiological
11.
J Lipid Res ; 57(11): 2051-2060, 2016 11.
Article in English | MEDLINE | ID: mdl-27623847

ABSTRACT

N-Acylphosphatidylethanolamines (NAPEs) are a class of glycerophospholipids, which are known as precursors for different bioactive N-acylethanolamines. We previously reported that phospholipase A/acyltransferase-1 (PLAAT-1), which was originally found in mammals as a tumor suppressor, catalyzes N-acylation of phosphatidylethanolamines to form NAPEs. However, recent online database suggested the presence of an uncharacterized isoform of PLAAT-1 with an extra sequence at the N terminus. In the present study, we examined the occurrence, intracellular localization, and catalytic properties of this longer isoform, as well as the original shorter isoform from humans and mice. Our results showed that human tissues express the longer isoform but not the short isoform at all. In contrast, mice expressed both isoforms with different tissue distribution. Unlike the cytoplasmic localization of the shorter isoform, the long isoform was found in both cytoplasm and nucleus, inferring that the extra sequence harbors a nuclear localization signal. As assayed with purified proteins, neither isoform required calcium for full activity. Moreover, the overexpression of each isoform remarkably increased cellular NAPE levels. These results conclude that the new long isoform of PLAAT-1 is a calcium-independent N-acyltransferase existing in both cytoplasm and nucleus and suggest a possible formation of NAPEs in various membrane structures including nuclear membrane. J. Lipid Res 2016. 57: 2051-2060.


Subject(s)
Acyltransferases/genetics , Phosphatidylethanolamines/biosynthesis , Phospholipases A1/genetics , Protein Isoforms/biosynthesis , Acylation , Acyltransferases/chemistry , Amino Acid Sequence/genetics , Animals , COS Cells , Calcium/metabolism , Catalysis , Cell Nucleus/enzymology , Chlorocebus aethiops , Cytoplasm/enzymology , Endocannabinoids/chemistry , Endocannabinoids/genetics , Gene Expression Regulation, Enzymologic , Glycerophospholipids/chemistry , Glycerophospholipids/genetics , Humans , Mice , Phosphatidylethanolamines/chemistry , Phospholipases A1/chemistry , Protein Isoforms/chemistry , Protein Isoforms/genetics
12.
Biochim Biophys Acta ; 1861(9 Pt A): 1058-1065, 2016 09.
Article in English | MEDLINE | ID: mdl-27317427

ABSTRACT

Mammalian cells maintain the glycerophospholipid (GPL) compositions of their membranes nearly constant. To achieve this, GPL synthesis and degradation must be coordinated. There is strong evidence that A-type phospholipases (PLAs) are key players in homeostatic degradation of GPLs, but the identities of the PLAs involved have not been established. However, some members of the Patatin-like phospholipase domain-containing proteins (PNPLAs) have been implicated. Accordingly, we knocked down all the PNPLAs significantly expressed in human HeLa cells using RNA interference and then determined whether the turnover of the major glycerophospholipids is affected by using mass spectrometry and metabolic labeling with stable isotope-labeled precursors. Knockdown of PNPLA9, PNPLA6 or PNPLA4 significantly (30-50%) reduced the turnover of phosphatidylcholine, -ethanolamine and -serine. In a notable contrast, turnover of phosphatidylinositol was not significantly affected by the knockdown of any PNPLA. Depletion of PNPLA9 and PNPLA4 also inhibited G0/G1 to S cell cycle progression, which could thus be regulated by GPL turnover. These results strongly suggest that PNPLA9, -6 and -4 play a key role in GPL turnover and homeostasis in human cells. A hypothetical model suggesting how these enzymes could recognize the relative concentration of the different GPLs is proposed.


Subject(s)
Glycerophospholipids/genetics , Lipase/genetics , Phospholipases/genetics , Cell Cycle/genetics , Cell Membrane/enzymology , Cell Membrane/genetics , Gene Expression Regulation , Gene Knockdown Techniques , Glycerophospholipids/metabolism , HeLa Cells , Homeostasis , Humans , Lipase/biosynthesis , Phosphatidylcholines/biosynthesis , Phospholipases/biosynthesis , Phospholipases/metabolism
13.
Food Funct ; 7(5): 2469-78, 2016 May 18.
Article in English | MEDLINE | ID: mdl-27161951

ABSTRACT

With the current changes in diet and living habits, obesity has become a global health problem. Thus, the weight-reducing function of tea has attracted considerable attention. This study investigated the anti-obesity effect and the mechanism of black tea (BT) polyphenols and polysaccharides in male Sprague-Dawley rats. The BT polyphenols and polysaccharides reduced the body weight, Lee's index, visceral fat weight, and fat cell size but improved the biochemical profile and increased the fecal fatty acid content, thereby preventing high-fat diet-induced obesity. A gene expression profile array was used to screen eight upregulated and five downregulated differentially expressed genes that affect fat metabolic pathways, such as glycerolipid and glycerophospholipid metabolism, fatty acid degradation, glycolysis and gluconeogenesis, bile and pancreatic secretion, the insulin signaling pathway, and steroid hormone secretion. The BT polyphenols and polysaccharides suppressed the formation and accumulation of fat and promoted its decomposition to prevent obesity.


Subject(s)
Body Composition/drug effects , Diet, High-Fat/adverse effects , Fatty Acids/metabolism , Feces/chemistry , Plant Extracts/therapeutic use , Polyphenols/therapeutic use , Polysaccharides/therapeutic use , Tea/chemistry , Adipose Tissue/pathology , Administration, Oral , Animals , Bile/metabolism , Blood Chemical Analysis , Body Weight/drug effects , Cell Size/drug effects , Eating , Fatty Acids/analysis , Fatty Acids/genetics , Gene Expression Regulation , Gluconeogenesis/genetics , Glycerophospholipids/genetics , Glycerophospholipids/metabolism , Glycolysis/genetics , Insulin/metabolism , Intra-Abdominal Fat/drug effects , Liver/chemistry , Liver/pathology , Male , Obesity/drug therapy , Plant Extracts/chemistry , Polyphenols/chemistry , Polyphenols/pharmacology , Polysaccharides/chemistry , Rats , Rats, Sprague-Dawley , Transcriptome
14.
Nucleic Acids Res ; 43(22): 10673-88, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26323317

ABSTRACT

The NF-κB-inducible Staphylococcal nuclease and tudor domain-containing 1 gene (SND1) encodes a coactivator involved in inflammatory responses and tumorigenesis. While SND1 is known to interact with certain transcription factors and activate client gene expression, no comprehensive mapping of SND1 target genes has been reported. Here, we have approached this question by performing ChIP-chip assays on human hepatoma HepG2 cells and analyzing SND1 binding modulation by proinflammatory TNFα. We show that SND1 binds 645 gene promoters in control cells and 281 additional genes in TNFα-treated cells. Transcription factor binding site analysis of bound probes identified motifs for established partners and for novel transcription factors including HSF, ATF, STAT3, MEIS1/AHOXA9, E2F and p300/CREB. Major target genes were involved in gene expression and RNA metabolism regulation, as well as development and cellular metabolism. We confirmed SND1 binding to 21 previously unrecognized genes, including a set of glycerolipid genes. Knocking-down experiments revealed that SND1 deficiency compromises the glycerolipid gene reprogramming and lipid phenotypic responses to TNFα. Overall, our findings uncover an unexpected large set of potential SND1 target genes and partners and reveal SND1 to be a determinant downstream effector of TNFα that contributes to support glycerophospholipid homeostasis in human hepatocellular carcinoma during inflammation.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Tumor Necrosis Factor-alpha/physiology , Binding Sites , Carcinoma, Hepatocellular/metabolism , Chromatin Immunoprecipitation , Endonucleases , Gene Expression Regulation, Neoplastic , Glycerophospholipids/genetics , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Nuclear Proteins/physiology , Oligonucleotide Array Sequence Analysis , Transcription Factors/metabolism
15.
J Biol Chem ; 289(19): 13575-88, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24675078

ABSTRACT

Acute fatty acid (FA) exposure potentiates glucose-stimulated insulin secretion in ß cells through metabolic and receptor-mediated effects. We assessed the effect of fatty acids on the dynamics of the metabolome in INS-1 cells following exposure to [U-(13)C]glucose to assess flux through metabolic pathways. Metabolite profiling showed a fatty acid-induced increase in long chain acyl-CoAs that were rapidly esterified with glucose-derived glycerol-3-phosphate to form lysophosphatidic acid, mono- and diacylglycerols, and other glycerolipids, some implicated in augmenting insulin secretion. Glucose utilization and glycolytic flux increased, along with a reduction in the NADH/NAD(+) ratio, presumably by an increase in conversion of dihydroxyacetone phosphate to glycerol-3-phosphate. The fatty acid-induced increase in glycolysis also resulted in increases in tricarboxylic cycle flux and oxygen consumption. Inhibition of fatty acid activation of FFAR1/GPR40 by an antagonist decreased glycerolipid formation, attenuated fatty acid increases in glucose oxidation, and increased mitochondrial FA flux, as evidenced by increased acylcarnitine levels. Conversely, FFAR1/GPR40 activation in the presence of low FA increased flux into glycerolipids and enhanced glucose oxidation. These results suggest that, by remodeling glucose and lipid metabolism, fatty acid significantly increases the formation of both lipid- and TCA cycle-derived intermediates that augment insulin secretion, increasing our understanding of mechanisms underlying ß cell insulin secretion.


Subject(s)
Fatty Acids/metabolism , Glucose/metabolism , Glycerophospholipids/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Line , Fatty Acids/genetics , Glucose/genetics , Glycerophospholipids/genetics , Insulin/genetics , Insulin Secretion , Insulin-Secreting Cells/cytology , Lipid Metabolism/physiology , Metabolome , Oxidation-Reduction , Rats , Receptors, G-Protein-Coupled/genetics
16.
Biochim Biophys Acta ; 1821(9): 1256-68, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22178194

ABSTRACT

Alterations in lipid metabolism and in the lipid composition of cellular membranes are linked to the pathology of numerous diseases including cancer. However, the influence of oncogene expression on cellular lipid profile is currently unknown. In this work we analyzed changes in lipid profiles that are induced in the course of ERBB2-expression mediated premature senescence. As a model system we used MCF-7 breast cancer cells with doxycycline-inducible expression of NeuT, an oncogenic ERBB2 variant. Affymetrix gene array data showed NeuT-induced alterations in the transcription of many enzymes involved in lipid metabolism, several of which (ACSL3, CHPT1, PLD1, LIPG, MGLL, LDL and NPC1) could be confirmed by quantitative realtime PCR. A study of the glycerophospholipid and lyso-glycerophospholipid profiles, obtained by high performance liquid chromatography coupled to Fourier-transform ion cyclotron resonance-mass spectrometry revealed senescence-associated changes in numerous lipid species, including mitochondrial lipids. The most prominent changes were found in PG(34:1), PG(36:1) (increased) and LPE(18:1), PG(40:7) and PI(36:1) (decreased). Statistical analysis revealed a general trend towards shortened phospholipid acyl chains in senescence and a significant trend to more saturated acyl chains in the class of phosphatidylglycerol. Additionally, the cellular cholesterol content was elevated and accumulated in vacuoles in senescent cells. These changes were accompanied by increased membrane fluidity. In mitochondria, loss of membrane potential along with altered intracellular distribution was observed. In conclusion, we present a comprehensive overview of altered cholesterol and glycerophospholipid patterns in senescence, showing that predominantly mitochondrial lipids are affected and lipid species less susceptible to peroxidation are increased.


Subject(s)
Breast Neoplasms/metabolism , Cellular Senescence , Genes, erbB-2 , Glycerophospholipids/metabolism , Lipid Metabolism , Receptor, ErbB-2/biosynthesis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Glycerophospholipids/genetics , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Receptor, ErbB-2/genetics , Vacuoles/genetics , Vacuoles/metabolism , Vacuoles/pathology
17.
J Biol Chem ; 286(39): 33819-31, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21803774

ABSTRACT

AT1G78690, a gene found in Arabidopsis thaliana, has been reported to encode a N-acyltransferase that transfers an acyl chain from acyl-CoA to the headgroup of phosphatidylethanolamine (PE) to form N-acylphosphatidylethanolamine (N-acyl-PE). Our investigation suggests that At1g78690p is not a PE-dependent N-acyltransferase but is instead a lysoglycerophospholipid O-acyltransferase. We overexpressed AT1G78690 in Escherichia coli, extracted the cellular lipids, and identified the accumulating glycerophospholipid as acylphosphatidylglycerol (acyl-PG). Electrospray ionization quadrupole time-of-flight mass spectrometry (ESI-MS) analysis yielded [M - H](-) ions, corresponding by exact mass to acyl-PG rather than N-acyl-PE. Collision-induced dissociation mass spectrometry (MS/MS) yielded product ions consistent with acyl-PG. In addition, in vitro enzyme assays using both (32)P- and (14)C-radiolabeled substrates showed that AT1G78690 acylates 1-acyllysophosphatidylethanolamine (1-acyllyso-PE) and 1-acyllysophosphatidylglycerol (1-acyllyso-PG), but not PE or phosphatidylglycerol (PG), to form a diacylated product that co-migrates with PE and PG, respectively. We analyzed the diacylated product formed by AT1G78690 using a combination of base hydrolysis, phospholipase D treatment, ESI-MS, and MS/MS to show that AT1G78690 acylates the sn-2-position of 1-acyllyso-PE and 1-acyllyso-PG.


Subject(s)
Acyltransferases/chemistry , Arabidopsis Proteins/chemistry , Arabidopsis/enzymology , Acylation , Acyltransferases/genetics , Acyltransferases/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Glycerophospholipids/biosynthesis , Glycerophospholipids/chemistry , Glycerophospholipids/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization
18.
J Biol Chem ; 286(43): 37676-91, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21873652

ABSTRACT

Loss-of-function mutations in 1-acylglycerol-3-phosphate O-acyltransferase (AGPAT) 2 in humans and mice result in loss of both the white and brown adipose tissues from birth. AGPAT2 generates precursors for the synthesis of glycerophospholipids and triacylglycerols. Loss of adipose tissue, or lipodystrophy, results in hyperinsulinemia, diabetes mellitus, and severe hepatic steatosis. Here, we analyzed biochemical properties of human AGPAT2 and its close homolog, AGPAT1, and we studied their role in liver by transducing their expression via recombinant adenoviruses in Agpat2(-/-) mice. The in vitro substrate specificities of AGPAT1 and AGPAT2 are quite similar for lysophosphatidic acid and acyl-CoA. Protein homology modeling of both the AGPATs with glycerol-3-phosphate acyltransferase 1 (GPAT1) revealed that they have similar tertiary protein structure, which is consistent with their similar substrate specificities. When co-expressed, both isoforms co-localize to the endoplasmic reticulum. Despite such similarities, restoring AGPAT activity in liver by overexpression of either AGPAT1 or AGPAT2 in Agpat2(-/-) mice failed to ameliorate the hepatic steatosis. From these studies, we suggest that the role of AGPAT1 or AGPAT2 in liver lipogenesis is minimal and that accumulation of liver fat is primarily a consequence of insulin resistance and loss of adipose tissue in Agpat2(-/-) mice.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Adipose Tissue/enzymology , Endoplasmic Reticulum/enzymology , Fatty Liver/enzymology , Lipodystrophy/enzymology , Liver/enzymology , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , Adipose Tissue/pathology , Animals , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Fatty Liver/genetics , Fatty Liver/pathology , Glycerophospholipids/biosynthesis , Glycerophospholipids/genetics , HEK293 Cells , Humans , Insulin Resistance/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Lipodystrophy/genetics , Lipodystrophy/pathology , Liver/pathology , Mice , Mice, Knockout , Transduction, Genetic , Triglycerides/biosynthesis , Triglycerides/genetics
19.
PLoS Genet ; 7(2): e1001299, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21347278

ABSTRACT

During Alzheimer's Disease, sustained exposure to amyloid-ß42 oligomers perturbs metabolism of ether-linked glycerophospholipids defined by a saturated 16 carbon chain at the sn-1 position. The intraneuronal accumulation of 1-O-hexadecyl-2-acetyl-sn-glycerophosphocholine (C16:0 PAF), but not its immediate precursor 1-O-hexadecyl-sn-glycerophosphocholine (C16:0 lyso-PAF), participates in signaling tau hyperphosphorylation and compromises neuronal viability. As C16:0 PAF is a naturally occurring lipid involved in cellular signaling, it is likely that mechanisms exist to protect cells against its toxic effects. Here, we utilized a chemical genomic approach to identify key processes specific for regulating the sensitivity of Saccharomyces cerevisiae to alkyacylglycerophosphocholines elevated in Alzheimer's Disease. We identified ten deletion mutants that were hypersensitive to C16:0 PAF and five deletion mutants that were hypersensitive to C16:0 lyso-PAF. Deletion of YDL133w, a previously uncharacterized gene which we have renamed SRF1 (Spo14 Regulatory Factor 1), resulted in the greatest differential sensitivity to C16:0 PAF over C16:0 lyso-PAF. We demonstrate that Srf1 physically interacts with Spo14, yeast phospholipase D (PLD), and is essential for PLD catalytic activity in mitotic cells. Though C16:0 PAF treatment does not impact hydrolysis of phosphatidylcholine in yeast, C16:0 PAF does promote delocalization of GFP-Spo14 and phosphatidic acid from the cell periphery. Furthermore, we demonstrate that, similar to yeast cells, PLD activity is required to protect mammalian neural cells from C16:0 PAF. Together, these findings implicate PLD as a potential neuroprotective target capable of ameliorating disruptions in lipid metabolism in response to accumulating oligomeric amyloid-ß42.


Subject(s)
Glycerophospholipids/metabolism , Phospholipase D/metabolism , Platelet Activating Factor/analogs & derivatives , Platelet Activating Factor/toxicity , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Alzheimer Disease/metabolism , Cell Line , Glycerophospholipids/genetics , Humans , Lipid Metabolism/genetics , Mutation/genetics , Neurons/metabolism , Phospholipase D/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
20.
J Biol Chem ; 284(1): 1-5, 2009 Jan 02.
Article in English | MEDLINE | ID: mdl-18718904

ABSTRACT

Cell membranes contain several classes of glycerophospholipids, which have numerous structural and functional roles in the cells. Polyunsaturated fatty acids, including arachidonic acid and eicosapentaenoic acid, are located at the sn-2 (but not sn-1)-position of glycerophospholipids in an asymmetrical manner. Using acyl-CoAs as donors, glycerophospholipids are formed by a de novo pathway (Kennedy pathway) and modified by a remodeling pathway (Lands' cycle) to generate membrane asymmetry and diversity. Both pathways were reported in the 1950s. Whereas enzymes involved in the Kennedy pathway have been well characterized, including enzymes in the 1-acylglycerol-3-phosphate O-acyltransferase family, little is known about enzymes involved in the Lands' cycle. Recently, several laboratories, including ours, isolated enzymes working in the remodeling pathway. These enzymes were discovered not only in the 1-acylglycerol-3-phosphate O-acyltransferase family but also in the membrane-bound O-acyltransferase family. In this review, we summarize recent studies on cloning and characterization of lysophospholipid acyltransferases that contribute to membrane asymmetry and diversity.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/genetics , 1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Cell Membrane/genetics , Cell Membrane/metabolism , Glycerophospholipids/biosynthesis , Acyl Coenzyme A/genetics , Acyl Coenzyme A/metabolism , Animals , Fatty Acids, Unsaturated/genetics , Fatty Acids, Unsaturated/metabolism , Glycerophospholipids/genetics , Humans , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL