Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cell Death Dis ; 13(10): 893, 2022 10 22.
Article in English | MEDLINE | ID: mdl-36273194

ABSTRACT

Noninflammatory clearance of dying cells by professional phagocytes, termed efferocytosis, is fundamental in both homeostasis and inflammatory fibrosis disease but has not been confirmed to occur in chronic pancreatitis (CP). Here, we investigated whether efferocytosis constitutes a novel regulatory target in CP and its mechanisms. PRSS1 transgenic (PRSS1Tg) mice were treated with caerulein to mimic CP development. Phospholipid metabolite profiling and epigenetic assays were performed with PRSS1Tg CP models. The potential functions of Atp8b1 in CP model were clarified using Atp8b1-overexpressing adeno-associated virus, immunofluorescence, enzyme-linked immunosorbent assay(ELISA), and lipid metabolomic approaches. ATAC-seq combined with RNA-seq was then used to identify transcription factors binding to the Atp8b1 promoter, and ChIP-qPCR and luciferase assays were used to confirm that the identified transcription factor bound to the Atp8b1 promoter, and to identify the specific binding site. Flow cytometry was performed to analyze the proportion of pancreatic macrophages. Decreased efferocytosis with aggravated inflammation was identified in CP. The lysophosphatidylcholine (LPC) pathway was the most obviously dysregulated phospholipid pathway, and LPC and Atp8b1 expression gradually decreased during CP development. H3K27me3 ChIP-seq showed that increased Atp8b1 promoter methylation led to transcriptional inhibition. Atp8b1 complementation substantially increased the LPC concentration and improved CP outcomes. Bhlha15 was identified as a transcription factor that binds to the Atp8b1 promoter and regulates phospholipid metabolism. Our study indicates that the acinar Atp8b1/LPC pathway acts as an important "find-me" signal for macrophages and plays a protective role in CP, with Atp8b1 transcription promoted by the acinar cell-specific transcription factor Bhlha15. Bhlha15, Atp8b1, and LPC could be clinically translated into valuable therapeutic targets to overcome the limitations of current CP therapies.


Subject(s)
Adenosine Triphosphatases , Lysophosphatidylcholines , Macrophages , Pancreatitis, Chronic , Animals , Mice , Acinar Cells/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Ceruletide/toxicity , Histones/metabolism , Inflammation/metabolism , Lysophosphatidylcholines/genetics , Lysophosphatidylcholines/metabolism , Macrophages/metabolism , Pancreatitis, Chronic/chemically induced , Pancreatitis, Chronic/genetics , Pancreatitis, Chronic/metabolism , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism , Transcription Factors/metabolism
2.
Protein Eng Des Sel ; 32(1): 1-11, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31396631

ABSTRACT

Phospholipase D (PLD) is an enzyme widely used for enzymatic synthesis of structured phospholipids (PLs) with modified head groups. These PLs are mainly used as food supplements and liposome ingredients. Still, there is a need for an enzyme that discriminates between PLs and lysoPLs, for specific detection of lysoPLs in various specimens and enzymatic synthesis of certain PLs from a mixed substrate. To meet this demand, we aimed at altering sn-2 acyl chain recognition of a PLD, leading to a variant enzyme preferably reacting on lysoPLs, by protein engineering. Based on the crystal structure of Streptomyces antibioticus PLD, W166 was targeted for saturation mutagenesis due to its strong interaction with the sn-2 acyl chain of the PL. Screening result pointed at W166R and W166K PLDs to selectively react on lysophosphatidylcholine (lysoPC), while not on PC. These variants showed a negative correlation between activity and sn-2 chain length of PL substrates. This behavior was not observed in the wild-type (WT)-PLD. Kinetic analysis revealed that the W166R and W166K variants have 7-10 times higher preference to lysoPC compared to the WT-PLD. Additionally, W166R PLD showed detectable activity toward glycero-3-phosphocholine, unlike the WT-PLD. Applicability of the lysoPC-preferring PLD was demonstrated by detection of lysoPC in the mixed PC/lysoPC sample and by the synthesis of cyclic phosphatidic acid. Structure model analyses supported the experimental findings and provided a basis for the structure model-based hypothesis on the observed behavior of the enzymes.


Subject(s)
Amino Acid Substitution , Bacterial Proteins/chemistry , Lysophosphatidylcholines/chemistry , Phospholipase D/chemistry , Streptomyces antibioticus/enzymology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Lysophosphatidylcholines/genetics , Lysophosphatidylcholines/metabolism , Mutagenesis, Site-Directed , Mutation, Missense , Phospholipase D/genetics , Phospholipase D/metabolism , Streptomyces antibioticus/genetics , Substrate Specificity/genetics
3.
J Biol Chem ; 294(16): 6214-6226, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30782848

ABSTRACT

Lysophospholipids (LPLs) are important lipid-signaling molecules in plants, of which lysophosphatidylcholine (lysoPC) is one of the most well-characterized LPLs, having important roles in plant stress responses. It is broken down by lysophospholipases, but the molecular mechanism involved in lysoPC degradation is unclear. Recombinant Arabidopsis thaliana ACYL-CoA-BINDING PROTEIN2 (AtACBP2) has been reported to bind lysoPC via its acyl-CoA-binding domain and also LYSOPHOSPHOLIPASE 2 (AtLYSOPL2) via its ankyrin repeats in vitro To investigate the interactions of AtACBP2 with AtLYSOPL2 and lysoPC in more detail, we conducted isothermal titration calorimetry with AtACBP270-354, an AtACBP2 derivative consisting of amino acids 70-354, containing both the acyl-CoA-binding domain and ankyrin repeats. We observed that the interactions of AtACBP270-354 with AtLYSOPL2 and lysoPC were both endothermic, favored by solvation entropy and opposed by enthalpy, with dissociation constants in the micromolar range. Of note, three AtLYSOPL2 catalytic triad mutant proteins (S147A, D268A, and H298A) bound lysoPC only weakly, with an exothermic burst and dissociation constants in the millimolar range. Furthermore, the binding affinity of lysoPC-premixed AtACBP270-354 to AtLYSOPL2 was 10-fold higher than that of AtACBP270-354 alone to AtLYSOPL2. We conclude that AtACBP2 may play a role in facilitating a direct interaction between AtLYSOPL2 and lysoPC. Our results suggest that AtACBP270-354 probably binds to lysoPC through a hydrophobic interface that enhances a hydrotropic interaction of AtACBP270-354 with AtLYSOPL2 and thereby facilitates AtLYSOPL2's lysophospholipase function.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis/enzymology , Carrier Proteins/chemistry , Lysophosphatidylcholines/chemistry , Lysophospholipase/chemistry , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Entropy , Hydrophobic and Hydrophilic Interactions , Lysophosphatidylcholines/genetics , Lysophosphatidylcholines/metabolism , Lysophospholipase/genetics , Lysophospholipase/metabolism , Protein Binding , Signal Transduction
4.
J Proteome Res ; 18(3): 1133-1144, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30706713

ABSTRACT

Hyperlipidemia, characterized by high serum lipids, is a risk factor for cardiovascular disease. Recent studies have identified an important role for celastrol, a proteasome inhibitor isolated from Tripterygium wilfordii Hook. F., in obesity-related metabolic disorders. However, the exact influences of celastrol on lipid metabolism remain largely unknown. Celastrol inhibited the terminal differentiation of 3T3-L1 adipocytes and decreased the levels of triglycerides in wild-type mice. Lipidomics analysis revealed that celastrol increased the metabolism of lysophosphatidylcholines (LPCs), phosphatidylcholines (PCs), sphingomyelins (SMs), and phosphatidylethanolamines (PEs). Further, celastrol reversed the tyloxapol-induced hyperlipidemia induced associated with increased plasma LPCs, PCs, SMs, and ceramides (CMs). Among these lipids, LPC(16:0), LPC(18:1), PC(22:2/15:0), and SM(d18:1/22:0) were also decreased by celastrol in cultured 3T3-L1 adipocytes, mice, and tyloxapol-treated mice. The mRNAs encoded by hepatic genes associated with lipid synthesis and catabolism, including Lpcat1, Pld1, Smpd3, and Sptc2, were altered in tyloxapol-induced hyperlipidemia, and significantly recovered by celastrol treatment. The effect of celastrol on lipid metabolism was significantly reduced in Fxr-null mice, resulting in decreased Cers6 and Acer2 mRNAs compared to wild-type mice. These results establish that FXR was responsible in part for the effects of celastrol in controlling lipid metabolism and contributing to the recovery of aberrant lipid metabolism in obesity-related metabolic disorders.


Subject(s)
Hyperlipidemias/drug therapy , Lipid Metabolism/drug effects , Proteasome Inhibitors/pharmacology , Triterpenes/pharmacology , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Cell Differentiation/drug effects , Diet, High-Fat , Gene Expression Regulation/drug effects , Humans , Hyperlipidemias/chemically induced , Hyperlipidemias/genetics , Liver/drug effects , Liver/metabolism , Lysophosphatidylcholines/genetics , Mice , Pentacyclic Triterpenes , Phosphatidylcholines/genetics , Phosphatidylethanolamines/genetics , Phospholipase D/genetics , Polyethylene Glycols/toxicity , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelins/genetics
5.
J Recept Signal Transduct Res ; 38(4): 367-371, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30396320

ABSTRACT

Lysophosphatidic acid (LPA) is a simple biological lipid and mediates several biological functions with LPA receptors (LPA1 to LPA6). In the present study, to assess whether LPA receptors promote cell-invasive activity of pancreatic cancer cells, highly invasion PANC-R9 cells were established from PANC-1 cells, using Matrigel-coated Cell Culture Insert. The cell-invasive activity of PANC-R9 cells was shown to be approximately 15 times higher than that of PANC-1 cells. LPAR1 expression level was markedly elevated in PANC-R9 cells in comparison with PANC-1 cells, while LPAR3 expression level was reduced. The cell-invasive activity of PANC-R9 cells was enhanced by LPA, but LPA had no impact on PANC-1 cell invasion. Before initiation of the cell invasion assay, PANC-R9 cells were pretreated with dioctanoylglycerol pyrophosphate (DGPP), an antagonist of LPA1/LPA3. The invasive activity of PANC-R9 cells was markedly suppressed by DGPP. Autotaxin (ATX) is a key enzyme that catalyzes the conversion of lysophosphatidylcholine (LPC) to LPA. ATX expression level was elevated in PANC-R9 cells compared with PANC-1 cells. In the presence of LPC, the cell motile activity of PANC-R9 cells was markedly stimulated. In contrast, LPC did not affect the cell motile activity of PANC-1 cells. PANC-R9 cell motility was inhibited by an ATX inhibitor, PF-8380. These results suggest that LPA signaling via LPA1 is a potent molecular target for the regulation of tumor progression in PANC-1 cells.


Subject(s)
Lysophosphatidylcholines/metabolism , Pancreatic Neoplasms/drug therapy , Phosphoric Diester Hydrolases/genetics , Receptors, Lysophosphatidic Acid/genetics , Benzoxazoles/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , Humans , Lysophosphatidylcholines/genetics , Neoplasm Invasiveness/genetics , Pancreatic Neoplasms/genetics , Phosphatidic Acids/metabolism , Piperazines/pharmacology , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction/drug effects
6.
Steroids ; 139: 28-34, 2018 11.
Article in English | MEDLINE | ID: mdl-30217786

ABSTRACT

Lysophosphatidylcholine (LPC), a major component of oxidized low-density lipoprotein, is associated with atherosclerosis, obesity, stroke, and cancer. However, the direction and mechanism of this relationship remains unclear. In this study, we conducted RNA profiling in THP-1 derived macrophages treated with LPC and uncovered a relationship between LPC and the cholesterol biosynthesis pathway. Principal component analysis (PCA) of RNA profiling showed that untreated THP-1 cells and those treated with 10, 20, or 40 µM LPC were distinctly distributed. Functional annotation revealed that LPC affected the expression of genes involved in cytokine-cytokine receptor interaction, TNF signaling, and MAPK signaling. Interestingly, LPC also altered the expression of 11 genes involved in cholesterol synthesis such as those in terpenoid backbone biosynthesis and steroid biosynthesis pathways. This increased gene expression occurred in a dose-dependent manner in response to LPC treatment. Especially, LPC with saturated acyl groups enhanced the expression of these genes compared to LPC with unsaturated acyl groups, and similar results were shown in response to saturated and unsaturated free fatty acids. Our findings demonstrate that LPCs with saturated acyl groups induce the expression of genes involved in cholesterol biosynthesis and may have implications for cholesterol related diseases.


Subject(s)
Atherosclerosis/genetics , Cholesterol/biosynthesis , Lipoproteins, LDL/biosynthesis , Lysophosphatidylcholines/pharmacology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cholesterol/genetics , Gene Expression/drug effects , Humans , Lipoproteins, LDL/genetics , Lysophosphatidylcholines/genetics , Macrophages/metabolism , Mitogen-Activated Protein Kinase Kinases/genetics , Principal Component Analysis , Signal Transduction/drug effects
7.
J Lipid Res ; 57(10): 1758-1770, 2016 10.
Article in English | MEDLINE | ID: mdl-27049024

ABSTRACT

The accumulation of lipids is a histologic and biochemical hallmark of obesity-associated nonalcoholic fatty liver disease (NAFLD). A subset of NALFD patients develops progressive liver disease, termed nonalcoholic steatohepatitis, which is characterized by hepatocellular apoptosis and innate immune system-mediated inflammation. These responses are orchestrated by signaling pathways that can be activated by lipids, directly or indirectly. In this review, we discuss palmitate- and lysophosphatidylcholine (LPC)-induced upregulation of p53-upregulated modulator of apoptosis and cell-surface expression of the death receptor TNF-related apoptosis-inducing ligand receptor 2. Next, we review the activation of stress-induced kinases, mixed lineage kinase 3, and c-Jun N-terminal kinase, and the activation of endoplasmic reticulum stress response and its downstream proapoptotic effector, CAAT/enhancer binding homologous protein, by palmitate and LPC. Moreover, the activation of these stress signaling pathways is linked to the release of proinflammatory, proangiogenic, and profibrotic extracellular vesicles by stressed hepatocytes. This review discusses the signaling pathways induced by lethal and sublethal lipid overload that contribute to the pathogenesis of NAFLD.


Subject(s)
Hepatocytes/metabolism , MAP Kinase Signaling System , Non-alcoholic Fatty Liver Disease/metabolism , Stress, Physiological , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Hepatocytes/pathology , Humans , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Lysophosphatidylcholines/genetics , Lysophosphatidylcholines/metabolism , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Palmitates/metabolism , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Up-Regulation , Mitogen-Activated Protein Kinase Kinase Kinase 11
8.
J Biol Chem ; 291(20): 10501-14, 2016 May 13.
Article in English | MEDLINE | ID: mdl-27008858

ABSTRACT

Eye photoreceptor membrane discs in outer rod segments are highly enriched in the visual pigment rhodopsin and the ω-3 fatty acid docosahexaenoic acid (DHA). The eye acquires DHA from blood, but transporters for DHA uptake across the blood-retinal barrier or retinal pigment epithelium have not been identified. Mfsd2a is a newly described sodium-dependent lysophosphatidylcholine (LPC) symporter expressed at the blood-brain barrier that transports LPCs containing DHA and other long-chain fatty acids. LPC transport via Mfsd2a has been shown to be necessary for human brain growth. Here we demonstrate that Mfsd2a is highly expressed in retinal pigment epithelium in embryonic eye, before the development of photoreceptors, and is the primary site of Mfsd2a expression in the eye. Eyes from whole body Mfsd2a-deficient (KO) mice, but not endothelium-specific Mfsd2a-deficient mice, were DHA-deficient and had significantly reduced LPC/DHA transport in vivo Fluorescein angiography indicated normal blood-retinal barrier function. Histological and electron microscopic analysis indicated that Mfsd2a KO mice exhibited a specific reduction in outer rod segment length, disorganized outer rod segment discs, and mislocalization of and reduction in rhodopsin early in postnatal development without loss of photoreceptors. Minor photoreceptor cell loss occurred in adult Mfsd2a KO mice, but electroretinography indicated visual function was normal. The developing eyes of Mfsd2a KO mice had activated microglia and up-regulation of lipogenic and cholesterogenic genes, likely adaptations to loss of LPC transport. These findings identify LPC transport via Mfsd2a as an important pathway for DHA uptake in eye and for development of photoreceptor membrane discs.


Subject(s)
Docosahexaenoic Acids/metabolism , Lysophosphatidylcholines/metabolism , Membrane Transport Proteins/metabolism , Photoreceptor Cells/metabolism , Angiography , Animals , Biological Transport, Active/physiology , Docosahexaenoic Acids/genetics , Lysophosphatidylcholines/genetics , Membrane Transport Proteins/genetics , Mice , Mice, Knockout , Microglia/metabolism , Optical Imaging , Symporters , Up-Regulation
9.
Proc Natl Acad Sci U S A ; 113(8): 2110-5, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26858457

ABSTRACT

Lipid oxidation products, including lysophosphatidylcholine (lysoPC), activate canonical transient receptor potential 6 (TRPC6) channels leading to inhibition of endothelial cell (EC) migration in vitro and delayed EC healing of arterial injuries in vivo. The precise mechanism through which lysoPC activates TRPC6 channels is not known, but calmodulin (CaM) contributes to the regulation of TRPC channels. Using site-directed mutagenesis, cDNAs were generated in which Tyr(99) or Tyr(138) of CaM was replaced with Phe, generating mutant CaM, Phe(99)-CaM, or Phe(138)-CaM, respectively. In ECs transiently transfected with pcDNA3.1-myc-His-Phe(99)-CaM, but not in ECs transfected with pcDNA3.1-myc-His-Phe(138)-CaM, the lysoPC-induced TRPC6-CaM dissociation and TRPC6 externalization was disrupted. Also, the lysoPC-induced increase in intracellular calcium concentration was inhibited in ECs transiently transfected with pcDNA3.1-myc-His-Phe(99)-CaM. Blocking phosphorylation of CaM at Tyr(99) also reduced CaM association with the p85 subunit and subsequent activation of phosphatidylinositol 3-kinase (PI3K). This prevented the increase in phosphatidylinositol (3,4,5)-trisphosphate (PIP3) and the translocation of TRPC6 to the cell membrane and reduced the inhibition of EC migration by lysoPC. These findings suggest that lysoPC induces CaM phosphorylation at Tyr(99) by a Src family kinase and that phosphorylated CaM activates PI3K to produce PIP3, which promotes TRPC6 translocation to the cell membrane.


Subject(s)
Calcium Signaling/physiology , Calmodulin/metabolism , Cell Membrane/metabolism , Cell Movement/physiology , Endothelial Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , TRPC Cation Channels/metabolism , Animals , Calcium/metabolism , Calmodulin/genetics , Cattle , Cell Membrane/genetics , Endothelial Cells/cytology , Enzyme Activation/physiology , Humans , Lysophosphatidylcholines/genetics , Lysophosphatidylcholines/metabolism , Phosphatidylinositol 3-Kinases/genetics , Protein Transport/physiology , TRPC Cation Channels/genetics , TRPC6 Cation Channel
10.
Am J Respir Cell Mol Biol ; 53(1): 33-41, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25780943

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a destructive lung disease affecting women. LAM is caused by mutations in the tuberous sclerosis complex (TSC) genes. The TSC protein complex inhibits the mechanistic/mammalian target of rapamycin complex 1 (mTORC1), which is a master regulator of cellular metabolism. Using mass spectrometry-based lipid profiling, we analyzed plasma from patients with LAM and discovered elevated levels of four lysophosphatidylcholine (LPC) species (C16:0, C18:0, C18:1, and C20:4) compared with those in healthy control women. To investigate whether these lipids are generated in a TSC2-dependent manner, we profiled in vitro preclinical models of TSC/LAM and found significant LPC accumulation in TSC2-deficient cells relative to TSC2-expressing control cells. These lysoglycerophospholipid changes occurred alongside changes in other phospholipid and neutral lipid species. Treatment with rapamycin or torin1 or down-regulation of sterol regulatory element-binding protein (SREBP), a lipogenic transcription factor, did not suppress LPC in TSC2-deficient cells. Inhibition of distinct isoforms of phospholipase A2 decreased the proliferation of TSC2-deficient cells. Collectively, these results demonstrate that TSC2-deficient cells have enhanced choline phospholipid metabolism and reveal a novel function of the TSC proteins in choline lysoglycerophospholipid metabolism, with implications for disease pathogenesis and targeted therapeutic strategies.


Subject(s)
Lipid Metabolism , Lymphangioleiomyomatosis/metabolism , Lysophosphatidylcholines/biosynthesis , Tumor Suppressor Proteins/deficiency , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Female , Humans , Lymphangioleiomyomatosis/genetics , Lymphangioleiomyomatosis/pathology , Lysophosphatidylcholines/genetics , Mass Spectrometry , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Naphthyridines/pharmacology , Phospholipases A2/genetics , Phospholipases A2/metabolism , Rats , Sirolimus/pharmacology , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 2 Protein
11.
J Biol Chem ; 288(36): 26013-26026, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-23897815

ABSTRACT

Defective clearance of apoptotic cells is frequently associated with perpetuation of inflammatory conditions. Our results show a rapid activation of AMP-activated kinase (AMPK) in macrophages upon exposure to apoptotic cells or lysophosphatidylcholine, a specific phospholipid that is produced and released from dying cells. AMPK activation resulted from inhibition of mitochondrial oxygen consumption and ATP production and further depended on Ca(2+) mobilization and mitochondrial reactive oxygen species generation. Once activated, AMPK increased microtubule synthesis and chemokinesis and provided adaptation to energy demand during tracking and engulfment. Uptake of apoptotic cells was increased in lungs of mice that received lysophosphatidylcholine. Furthermore, inhibition of AMPK diminished clearance of apoptotic thymocytes in vitro and in dexamethasone-treated mice. Taken together, we conclude that the mitochondrial AMPK axis is a sensor and enhancer of tracking and removal of apoptotic cell, processes crucial to resolution of inflammatory conditions and a return to tissue homeostasis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Apoptosis/physiology , Macrophages, Peritoneal/metabolism , Mitochondria/metabolism , AMP-Activated Protein Kinases/genetics , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Animals , Cell Movement/physiology , Enzyme Activation/physiology , Lung/cytology , Lung/metabolism , Lysophosphatidylcholines/genetics , Lysophosphatidylcholines/metabolism , Macrophages, Peritoneal/cytology , Male , Mice , Mitochondria/genetics , Oxygen Consumption/physiology , Thymocytes/cytology , Thymocytes/metabolism
12.
J Biol Chem ; 287(18): 14880-95, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22389508

ABSTRACT

Calcium-independent phospholipase A(2)γ (iPLA(2)γ) (PNPLA8) is the predominant phospholipase activity in mammalian mitochondria. However, the chemical mechanisms that regulate its activity are unknown. Here, we utilize iPLA(2)γ gain of function and loss of function genetic models to demonstrate the robust activation of iPLA(2)γ in murine myocardial mitochondria by Ca(2+) or Mg(2+) ions. Calcium ion stimulated the production of 2-arachidonoyl-lysophosphatidylcholine (2-AA-LPC) from 1-palmitoyl-2-[(14)C]arachidonoyl-sn-glycero-3-phosphocholine during incubations with wild-type heart mitochondrial homogenates. Furthermore, incubation of mitochondrial homogenates from transgenic myocardium expressing iPLA(2)γ resulted in 13- and 25-fold increases in the initial rate of radiolabeled 2-AA-LPC and arachidonic acid (AA) production, respectively, in the presence of calcium ion. Mass spectrometric analysis of the products of calcium-activated hydrolysis of endogenous mitochondrial phospholipids in transgenic iPLA(2)γ mitochondria revealed the robust production of AA, 2-AA-LPC, and 2-docosahexaenoyl-LPC that was over 10-fold greater than wild-type mitochondria. The mechanism-based inhibitor (R)-(E)-6-(bromomethylene)-3-(1-naphthalenyl)-2H-tetrahydropyran-2-one (BEL) (iPLA(2)γ selective), but not its enantiomer, (S)-BEL (iPLA(2)ß selective) or pyrrolidine (cytosolic PLA(2)α selective), markedly attenuated Ca(2+)-dependent fatty acid release and polyunsaturated LPC production. Moreover, Ca(2+)-induced iPLA(2)γ activation was accompanied by the production of downstream eicosanoid metabolites that were nearly completely ablated by (R)-BEL or by genetic ablation of iPLA(2)γ. Intriguingly, Ca(2+)-induced iPLA(2)γ activation was completely inhibited by long-chain acyl-CoA (IC(50) ∼20 µm) as well as by a nonhydrolyzable acyl-CoA thioether analog. Collectively, these results demonstrate that mitochondrial iPLA(2)γ is activated by divalent cations and inhibited by acyl-CoA modulating the generation of biologically active metabolites that regulate mitochondrial bioenergetic and signaling functions.


Subject(s)
Arachidonic Acid/metabolism , Calcium/metabolism , Group VI Phospholipases A2/metabolism , Magnesium/metabolism , Mitochondria, Heart/enzymology , Animals , Arachidonic Acid/genetics , Cations, Divalent/metabolism , Enzyme Activation/drug effects , Group VI Phospholipases A2/antagonists & inhibitors , Group VI Phospholipases A2/genetics , Lysophosphatidylcholines/genetics , Lysophosphatidylcholines/metabolism , Mice , Mice, Knockout , Mitochondria, Heart/genetics , Models, Genetic , Naphthalenes/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Pyrones/pharmacology
13.
J Biol Chem ; 283(48): 33483-97, 2008 Nov 28.
Article in English | MEDLINE | ID: mdl-18801741

ABSTRACT

Among the many mammalian secreted phospholipase A2 (sPLA2) enzymes, PLA2G3 (group III secreted phospholipase A2) is unique in that it possesses unusual N- and C-terminal domains and in that its central sPLA2 domain is homologous to bee venom PLA2 rather than to other mammalian sPLA2s. To elucidate the in vivo actions of this atypical sPLA2, we generated transgenic (Tg) mice overexpressing human PLA2G3. Despite marked increases in PLA2 activity and mature 18-kDa PLA2G3 protein in the circulation and tissues, PLA2G3 Tg mice displayed no apparent abnormality up to 9 months of age. However, alterations in plasma lipoproteins were observed in PLA2G3 Tg mice compared with control mice. In vitro incubation of low density (LDL) and high density (HDL) lipoproteins with several sPLA2s showed that phosphatidylcholine was efficiently converted to lysophosphatidylcholine by PLA2G3 as well as by PLA2G5 and PLA2G10, to a lesser extent by PLA2G2F, and only minimally by PLA2G2A and PLA2G2E. PLA2G3-modified LDL, like PLA2G5- or PLA2G10-treated LDL, facilitated the formation of foam cells from macrophages ex vivo. Accumulation of PLA2G3 was detected in the atherosclerotic lesions of humans and apoE-deficient mice. Furthermore, following an atherogenic diet, aortic atherosclerotic lesions were more severe in PLA2G3 Tg mice than in control mice on the apoE-null background, in combination with elevated plasma lysophosphatidylcholine and thromboxane A2 levels. These results collectively suggest a potential functional link between PLA2G3 and atherosclerosis, as has recently been proposed for PLA2G5 and PLA2G10.


Subject(s)
Atherosclerosis/enzymology , Foam Cells/enzymology , Group III Phospholipases A2/metabolism , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Protein Processing, Post-Translational , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Bee Venoms/chemistry , Diet, Atherogenic , Foam Cells/pathology , Group III Phospholipases A2/chemistry , Group III Phospholipases A2/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Lipoproteins, HDL/genetics , Lipoproteins, LDL/genetics , Lysophosphatidylcholines/genetics , Lysophosphatidylcholines/metabolism , Mice , Mice, Transgenic , Phosphatidylcholines/genetics , Phosphatidylcholines/metabolism , Protein Processing, Post-Translational/genetics , Protein Structure, Tertiary/physiology , Sequence Homology, Amino Acid , Substrate Specificity/genetics
14.
J Biol Chem ; 283(12): 7776-89, 2008 Mar 21.
Article in English | MEDLINE | ID: mdl-18175805

ABSTRACT

Autotaxin is a type II ectonucleotide pyrophosphate phosphodiesterase enzyme. It has been recently discovered that it also has a lysophospholipase D activity. This enzyme probably provides most of the extracellular lysophosphatidic acid from lysophosphatidylcholine. The cloning and tissue distribution of the three isoforms (imaginatively called alpha, beta, and gamma) from human and mouse are reported in this study, as well as their tissue distribution by PCR in the human and mouse. The fate of the alpha isoform from human was also studied after purification and using mass spectrometry. Indeed, this particular isoform expresses the intron 12 in which a cleavage site is present, leading to a rapid catabolism of the isoform. For the human isoform gamma and the total autotaxin mRNA expression, quantitative PCR is presented in 21 tissues. The isoforms were expressed in two different hosts, insect cells and Chinese hamster ovary cells, and were highly purified. The characteristics of the six purified isoforms (pH and temperature dependence, K(m) and V(max) values, and their dependence on metal ions) are presented in this study. Their sensitivity to a small molecule inhibitor, hypericin, is also shown. Finally, the specificity of the isoforms toward a large family of lysophosphatidylcholines is reported. This study is the first complete description of the reported autotaxin isoforms.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Lysophosphatidylcholines/metabolism , Lysophospholipids/metabolism , Multienzyme Complexes/biosynthesis , Multienzyme Complexes/genetics , Phosphodiesterase I/biosynthesis , Phosphodiesterase I/genetics , Pyrophosphatases/biosynthesis , Pyrophosphatases/genetics , Animals , Anthracenes , Base Sequence , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Cricetulus , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/biosynthesis , Isoenzymes/genetics , Lysophosphatidylcholines/genetics , Lysophospholipids/genetics , Mice , Molecular Sequence Data , Multienzyme Complexes/antagonists & inhibitors , Organ Specificity/drug effects , Organ Specificity/physiology , Perylene/analogs & derivatives , Perylene/pharmacology , Phosphodiesterase I/antagonists & inhibitors , Phosphoric Diester Hydrolases/biosynthesis , Phosphoric Diester Hydrolases/genetics , Pyrophosphatases/antagonists & inhibitors , Substrate Specificity/drug effects , Substrate Specificity/physiology
15.
J Biol Chem ; 283(9): 5908-17, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18086666

ABSTRACT

Neuropathy target esterase (NTE) is a member of the family of patatin domain-containing proteins and exhibits phospholipase activity in brain and cultured cells. NTE was originally identified as target enzyme for organophosphorus compounds that cause a delayed paralyzing syndrome with degeneration of nerve axons. Here we show that the structurally related murine protein NTE-related esterase (NRE) is a potent lysophospholipase. The enzyme efficiently hydrolyzes sn-1 esters in lysophosphatidylcholine and lysophosphatidic acid. No lipase activity was observed when triacylglycerols, cholesteryl esters, retinyl esters, phosphatidylcholine, or monoacylglycerol were used as substrates. Although NTE is predominantly expressed in the nervous system, we found the highest NRE mRNA levels in testes, skeletal muscle, cardiac muscle, and adipose tissue. Induction of NRE mRNA concentrations in these tissues during fasting suggested a nutritional regulation of enzyme expression and, in accordance with this observation, insulin reduced NRE mRNA levels in a dose-dependent manner in 3T3-L1 adipocytes. A green fluorescent protein-NRE fusion protein colocalized to the endoplasmic reticulum and lipid droplets. Thus, NRE is a previously unrecognized ER- and lipid droplet-associated lysophospholipase. Regulation of enzyme expression by the nutritional status and insulin suggests a role of NRE in the catabolism of lipid precursors and/or mediators that affect energy metabolism in mammals.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Gene Expression Regulation, Enzymologic/physiology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Lipid Metabolism/physiology , Lysophospholipase/biosynthesis , 3T3-L1 Cells , Animals , Axons/enzymology , Brain/enzymology , Carboxylic Ester Hydrolases/genetics , Dose-Response Relationship, Drug , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/genetics , Fasting/physiology , Gene Expression Regulation, Enzymologic/drug effects , Lipid Metabolism/drug effects , Lysophosphatidylcholines/genetics , Lysophosphatidylcholines/metabolism , Lysophospholipase/genetics , Lysophospholipids/genetics , Lysophospholipids/metabolism , Male , Mice , Organ Specificity/physiology , Paralysis/enzymology , Paralysis/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Substrate Specificity/physiology , Syndrome
16.
J Biol Chem ; 282(51): 36853-61, 2007 Dec 21.
Article in English | MEDLINE | ID: mdl-17951629

ABSTRACT

We recently described a new route for the synthesis of phosphatidylethanolamine (PtdEtn) from exogenous lyso-PtdEtn, which we have termed the exogenous lysolipid metabolism (ELM) pathway. The ELM pathway for lyso-PtdEtn requires the action of plasma membrane P-type ATPases Dnf1p and Dnf2p and their requisite beta-subunit, Lem3p, for the active uptake of lyso-PtdEtn. In addition, the acyl-CoA-dependent acyltransferase, Ale1p, mediates the acylation of the imported lysolipid to form PtdEtn. We now report that these components of the lyso-PtdEtn ELM pathway are also active with lyso-1-acyl-2-hydroxyl-sn-glycero-3-phosphocholine (PtdCho) as a substrate. Lyso-PtdCho supports the growth of a choline auxotrophic pem1Delta pem2Delta strain. Uptake of radiolabeled lyso-PtdCho was impaired by the dnf2Delta and lem3Delta mutations. Introduction of a lem3Delta mutation into a pem1Delta pem2Delta background impaired the ability of the resulting strain to grow with lyso-PtdCho as the sole precursor of PtdCho. After import of lyso-PtdCho, the recently characterized lyso-PtdEtn acyltransferase, Ale1p, functioned as the sole lyso-PtdCho acyltransferase in yeast. A pem1Delta pem2Delta ale1Delta strain grew with lyso-PtdCho as a substrate but showed a profound reduction in PtdCho content when lyso-PtdCho was the only precursor of PtdCho. Ale1p acylates lyso-PtdCho with a preference for monounsaturated acyl-CoA species, and the specific LPCAT activity of Ale1p in yeast membranes is >50-fold higher than the basal rate of de novo aminoglycerophospholipid biosynthesis from phosphatidylserine synthase activity. In addition to lyso-PtdCho, lyso-PtdEtn, and lyso-phosphatidic acid, Ale1p was also active with lysophosphatidylserine, lysophosphatidylglycerol, and lysophosphatidylinositol as substrates. These results establish a new pathway for the net synthesis of PtdCho in yeast and provide new tools for the study of PtdCho synthesis, transport, and remodeling.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Adenosine Triphosphatases/metabolism , Cell Membrane/enzymology , Lysophosphatidylcholines/biosynthesis , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , ATP-Binding Cassette Transporters , Acylation , Adenosine Triphosphatases/genetics , Biological Transport, Active/physiology , Cell Membrane/genetics , Gene Deletion , Lysophosphatidylcholines/genetics , Lysophospholipids/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Phosphatidylethanolamine N-Methyltransferase/genetics , Phosphatidylethanolamine N-Methyltransferase/metabolism , Phosphatidylethanolamines/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Substrate Specificity/physiology
17.
J Biol Chem ; 282(42): 30562-9, 2007 Oct 19.
Article in English | MEDLINE | ID: mdl-17726007

ABSTRACT

The incorporation of unsaturated acyl chains into phospholipids during de novo synthesis is primarily mediated by the 1-acyl-sn-glycerol-3-phosphate acyltransferase reaction. In Saccharomyces cerevisiae, Slc1 has been shown to mediate this reaction, but distinct activity remains after its removal from the genome. To identify the enzyme that mediates the remaining activity, we performed synthetic genetic array analysis using a slc1Delta strain. One of the genes identified by the screen, LPT1, was found to encode for an acyltransferase that uses a variety of lysophospholipid species, including 1-acyl-sn-glycerol-3-phosphate. Deletion of LPT1 had a minimal effect on 1-acyl-sn-glycerol-3-phosphate acyltransferase activity, but overexpression increased activity 7-fold. Deletion of LPT1 abrogated the esterification of other lysophospholipids, and overexpression increased lysophosphatidylcholine acyltransferase activity 7-fold. The majority of this activity co-purified with microsomes. To test the putative role for this enzyme in selectively incorporating unsaturated acyl chains into phospholipids in vitro, substrate concentration series experiments were performed with the four acyl-CoA species commonly found in yeast. Although the saturated palmitoyl-CoA and stearoyl-CoA showed a lower apparent Km, the monounsaturated palmitoleoyl-CoA and oleoyl-CoA showed a higher apparent Vmax. Arachidonyl-CoA, although not abundant in yeast, also had a high apparent Vmax. Pulse-labeling of lpt1Delta strains showed a 30% reduction in [3H]oleate incorporation into phosphatidylcholine only. Therefore, Lpt1p, a member of the membrane-bound o-acyltransferase gene family, seems to work in conjunction with Slc1 to mediate the incorporation of unsaturated acyl chains into the sn-2 position of phospholipids.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Acyltransferases/metabolism , Cell Membrane/enzymology , Fatty Acids, Unsaturated/metabolism , Lysophosphatidylcholines/biosynthesis , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , Acyltransferases/genetics , Cell Membrane/genetics , Coenzymes/metabolism , Dyneins , Fatty Acids, Unsaturated/genetics , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Fungal/physiology , Kinetics , Lysophosphatidylcholines/genetics , Oligonucleotide Array Sequence Analysis , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...