Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Am J Physiol Endocrinol Metab ; 303(11): E1386-95, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23074238

ABSTRACT

Death of ß-cells due to apoptosis is an important contributor to ß-cell dysfunction in both type 1 and type 2 diabetes mellitus. Previously, we described participation of the Group VIA Ca(2+)-independent phospholipase A(2) (iPLA(2)ß) in apoptosis of insulinoma cells due to ER stress. To examine whether islet ß-cells are similarly susceptible to ER stress and undergo iPLA(2)ß-mediated apoptosis, we assessed the ER stress response in human pancreatic islets. Here, we report that the iPLA(2)ß protein is expressed predominantly in the ß-cells of human islets and that thapsigargin-induced ER stress promotes ß-cell apoptosis, as reflected by increases in activated caspase-3 in the ß-cells. Furthermore, we demonstrate that ER stress is associated with increases in islet iPLA(2)ß message, protein, and activity, iPLA(2)ß-dependent induction of neutral sphingomyelinase and ceramide accumulation, and subsequent loss of mitochondrial membrane potential. We also observe that basal activated caspase-3 increases with age, raising the possibility that ß-cells in older human subjects have a greater susceptibility to undergo apoptotic cell death. These findings reveal for the first time expression of iPLA(2)ß protein in human islet ß-cells and that induction of iPLA(2)ß during ER stress contributes to human islet ß-cell apoptosis. We hypothesize that modulation of iPLA(2)ß activity might reduce ß-cell apoptosis and this would be beneficial in delaying or preventing ß-cell dysfunction associated with diabetes.


Subject(s)
Apoptosis/physiology , Endoplasmic Reticulum/metabolism , Insulin-Secreting Cells/enzymology , Islets of Langerhans/enzymology , Phospholipases A2, Calcium-Independent/metabolism , Adult , Cells, Cultured , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/pathology , Enzyme Inhibitors/pharmacology , Female , Humans , In Vitro Techniques , Insulin-Secreting Cells/drug effects , Islets of Langerhans/cytology , Male , Phospholipases A2, Calcium-Independent/drug effects , Thapsigargin/pharmacology
2.
J Biol Chem ; 285(9): 6693-705, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20032468

ABSTRACT

Our recent studies indicate that endoplasmic reticulum (ER) stress causes INS-1 cell apoptosis by a Ca(2+)-independent phospholipase A(2) (iPLA(2)beta)-mediated mechanism that promotes ceramide generation via sphingomyelin hydrolysis and subsequent activation of the intrinsic pathway. To elucidate the association between iPLA(2)beta and ER stress, we compared beta-cell lines generated from wild type (WT) and Akita mice. The Akita mouse is a spontaneous model of ER stress that develops hyperglycemia/diabetes due to ER stress-induced beta-cell apoptosis. Consistent with a predisposition to developing ER stress, basal phosphorylated PERK and activated caspase-3 are higher in the Akita cells than WT cells. Interestingly, basal iPLA(2)beta, mature SREBP-1 (mSREBP-1), phosphorylated Akt, and neutral sphingomyelinase (NSMase) are higher, relative abundances of sphingomyelins are lower, and mitochondrial membrane potential (DeltaPsi) is compromised in Akita cells, in comparison with WT cells. Exposure to thapsigargin accelerates DeltaPsi loss and apoptosis of Akita cells and is associated with increases in iPLA(2)beta, mSREBP-1, and NSMase in both WT and Akita cells. Transfection of Akita cells with iPLA(2)beta small interfering RNA, however, suppresses NSMase message, DeltaPsi loss, and apoptosis. The iPLA(2)beta gene contains a sterol-regulatory element, and transfection with a dominant negative SREBP-1 reduces basal mSREBP-1 and iPLA(2)beta in the Akita cells and suppresses increases in mSREBP-1 and iPLA(2)beta due to thapsigargin. These findings suggest that ER stress leads to generation of mSREBP-1, which can bind to the sterol-regulatory element in the iPLA(2)beta gene to promote its transcription. Consistent with this, SREBP-1, iPLA(2)beta, and NSMase messages in Akita mouse islets are higher than in WT islets.


Subject(s)
Diabetes Mellitus/etiology , Endoplasmic Reticulum/pathology , Phospholipases A2, Calcium-Independent/genetics , Sterol Regulatory Element Binding Protein 1/physiology , Transcription, Genetic , Animals , Apoptosis , Binding Sites , Humans , Insulin-Secreting Cells , Mice , Mice, Transgenic , Protein Binding , Stress, Physiological
3.
Arterioscler Thromb Vasc Biol ; 27(3): 690-6, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17185615

ABSTRACT

OBJECTIVE: 3-Hydroxy-3-methylglutaryl coenzyme A (HMG CoA) reductase inhibitors (statins) exhibit antithrombotic properties that are independent of reductions in circulating LDL cholesterol. We hypothesized that these antithrombotic properties are mediated by membrane alterations secondary to disrupted lipid metabolism. METHODS AND RESULTS: EA.hy926 cells were incubated in the presence of 1 micromol/L atorvastatin supplemented with fetal bovine serum or lipid-depleted serum mixtures. Lipid restriction alone had no effect on cell lipid composition but when atorvastatin was included, phosphatidylserine, sphingomyelin, and cholesterol were reduced by 50% while ceramide content decreased by 70%. These changes in lipid composition did not alter the association of decay accelerating factor or tissue factor with lipid rafts. Atorvastatin in combination with lipid restriction reduced factor VIIa/tissue factor activity by as much as 75% but did not alter tissue factor expression. Prothrombinase activity was reduced to an extent similar to factor VIIa/tissue factor. Mevalonic acid but not LDL reversed the observed changes in lipid content and prothrombinase activity induced by atorvastatin. These findings were confirmed in primary cells. CONCLUSIONS: Inhibition of HMG-CoA reductase limits exposure of phosphatidylserine at the cell surface by restricting the cellular pool of mevalonate-derived isoprenoids. This membrane alteration restricts the activity of proteolytic enzyme complexes that propagate the coagulation cascade.


Subject(s)
Factor VIIa/drug effects , Fibroblasts/drug effects , Heptanoic Acids/pharmacology , Hydroxymethylglutaryl CoA Reductases/drug effects , Phosphatidylserines/metabolism , Pyrroles/pharmacology , Thromboplastin/metabolism , Animals , Atorvastatin , Blotting, Western , Cattle , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Factor VIIa/metabolism , Fibroblasts/physiology , Humans , Hydroxymethylglutaryl CoA Reductases/metabolism , Sensitivity and Specificity , Thromboplastin/drug effects
4.
Biochim Biophys Acta ; 1580(1): 40-56, 2002 Jan 30.
Article in English | MEDLINE | ID: mdl-11923099

ABSTRACT

In the evolution of Type II diabetes, an initial period of hyper-fatty acidemia leads to an insulin secretory defect which triggers overt hyperglycemia and frank diabetes. The mechanism by which elevated free fatty acids contribute to beta-cell dysfunction, however, is not clearly understood. We recently reported that arachidonic acid (20:4) or linoleic acid (18:2) supplementations result in increases in abundances of long chain polyunsaturated fatty acids in INS-1 beta-cell membrane lipids, suggesting that beta-cells express desaturases that catalyze generation of unsaturated fatty acids. As expression of desaturases by beta-cells has not yet been addressed, we initiated studies to examine this issue using INS-1 beta-cells and find that they express messages for the Delta6-, stearoyl CoA-, and Delta5-desaturase. Supplementation of the INS-1 beta-cells with arachidonic acid leads to decreased expression of all three desaturases, presumably in response to the decreased need for endogenous generation of unsaturated fatty acids. In contrast, linoleic acid supplementation promoted minimal changes in the three desaturases. These findings demonstrate for the first time that beta-cells express regulatable desaturases. Additionally, reverse transcriptase-polymerase chain reaction analyses reveal expression of the desaturases in native pancreatic islets. It might be speculated that long-term elevations in fatty acids can also adversely influence desaturase activity in beta-cells and affect PUFA composition in beta-cell membranes contributing to beta-cell membrane structural abnormalities and altered secretory function.


Subject(s)
Fatty Acid Desaturases/metabolism , Fatty Acids, Unsaturated/pharmacology , Islets of Langerhans/drug effects , Animals , Arachidonic Acid/pharmacology , Delta-5 Fatty Acid Desaturase , Dose-Response Relationship, Drug , Fatty Acid Desaturases/biosynthesis , Fatty Acid Desaturases/genetics , Islets of Langerhans/enzymology , Linoleic Acid/pharmacology , Linoleoyl-CoA Desaturase , Mass Spectrometry , Phosphatidylethanolamines/metabolism , Phosphorylcholine/metabolism , RNA, Messenger/analysis , Rats , Reverse Transcriptase Polymerase Chain Reaction , Stearoyl-CoA Desaturase/biosynthesis , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Tumor Cells, Cultured
5.
Endocrinology ; 155(9): 3352-64, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25004092

ABSTRACT

Type 1 diabetes (T1D) results from autoimmune destruction of islet ß-cells, but the underlying mechanisms that contribute to this process are incompletely understood, especially the role of lipid signals generated by ß-cells. Proinflammatory cytokines induce ER stress in ß-cells and we previously found that the Ca(2+)-independent phospholipase A2ß (iPLA2ß) participates in ER stress-induced ß-cell apoptosis. In view of reports of elevated iPLA2ß in T1D, we examined if iPLA2ß participates in cytokine-mediated islet ß-cell apoptosis. We find that the proinflammatory cytokine combination IL-1ß+IFNγ, induces: a) ER stress, mSREBP-1, and iPLA2ß, b) lysophosphatidylcholine (LPC) generation, c) neutral sphingomyelinase-2 (NSMase2), d) ceramide accumulation, e) mitochondrial membrane decompensation, f) caspase-3 activation, and g) ß-cell apoptosis. The presence of a sterol regulatory element in the iPLA2ß gene raises the possibility that activation of SREBP-1 after proinflammatory cytokine exposure contributes to iPLA2ß induction. The IL-1ß+IFNγ-induced outcomes (b-g) are all inhibited by iPLA2ß inactivation, suggesting that iPLA2ß-derived lipid signals contribute to consequential islet ß-cell death. Consistent with this possibility, ER stress and ß-cell apoptosis induced by proinflammatory cytokines are exacerbated in islets from RIP-iPLA2ß-Tg mice and blunted in islets from iPLA2ß-KO mice. These observations suggest that iPLA2ß-mediated events participate in amplifying ß-cell apoptosis due to proinflammatory cytokines and also that iPLA2ß activation may have a reciprocal impact on ER stress development. They raise the possibility that iPLA2ß inhibition, leading to ameliorations in ER stress, apoptosis, and immune responses resulting from LPC-stimulated immune cell chemotaxis, may be beneficial in preserving ß-cell mass and delaying/preventing T1D evolution.


Subject(s)
Apoptosis , Cytokines/immunology , Diabetes Mellitus, Type 1/enzymology , Group VI Phospholipases A2/immunology , Interferon-gamma/immunology , Interleukin-1beta/immunology , Islets of Langerhans/cytology , Adult , Animals , Cytokines/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/physiopathology , Endoplasmic Reticulum Stress , Female , Group VI Phospholipases A2/genetics , Humans , Interferon-gamma/genetics , Interleukin-1beta/genetics , Islets of Langerhans/enzymology , Islets of Langerhans/immunology , Male , Mice , Mice, Knockout , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/immunology
6.
Islets ; 5(1): 29-44, 2013.
Article in English | MEDLINE | ID: mdl-23411472

ABSTRACT

ß-cell apoptosis is a significant contributor to ß-cell dysfunction in diabetes and ER stress is among the factors that contributes to ß-cell death. We previously identified that the Ca²âº-independent phospholipase A2ß (iPLA2ß), which in islets is localized in ß-cells, participates in ER stress-induced ß-cell apoptosis. Here, direct assessment of iPLA2ß role was made using ß-cell-specific iPLA2ß overexpressing (RIP-iPLA2ß-Tg) and globally iPLA2ß-deficient (iPLA2ß-KO) mice. Islets from Tg, but not KO, express higher islet iPLA2ß and neutral sphingomyelinase, decrease in sphingomyelins, and increase in ceramides, relative to WT group. ER stress induces iPLA2ß, ER stress factors, loss of mitochondrial membrane potential (∆Ψ), caspase-3 activation, and ß-cell apoptosis in the WT and these are all amplified in the Tg group. Surprisingly, ß-cells apoptosis while reduced in the KO is higher than in the WT group. This, however, was not accompanied by greater caspase-3 activation but with larger loss of ∆Ψ, suggesting that iPLA2ß deficiency impacts mitochondrial membrane integrity and causes apoptosis by a caspase-independent manner. Further, autophagy, as reflected by LC3-II accumulation, is increased in Tg and decreased in KO, relative to WT. Our findings suggest that (1) iPLA2ß impacts upstream (UPR) and downstream (ceramide generation and mitochondrial) pathways in ß-cells and (2) both over- or under-expression of iPLA2ß is deleterious to the ß-cells. Further, we present for the first time evidence for potential regulation of autophagy by iPLA2ß in islet ß-cells. These findings support the hypothesis that iPLA2ß induction under stress, as in diabetes, is a key component to amplifying ß-cell death processes.


Subject(s)
Apoptosis , Autophagy , Endoplasmic Reticulum Stress , Gene Expression Regulation, Enzymologic , Group IV Phospholipases A2/metabolism , Insulin-Secreting Cells/metabolism , Animals , Apoptosis/drug effects , Autophagy/drug effects , Caspase 3/metabolism , Ceramides/metabolism , Diabetes Mellitus/enzymology , Diabetes Mellitus/metabolism , Endoplasmic Reticulum Stress/drug effects , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Group IV Phospholipases A2/biosynthesis , Group IV Phospholipases A2/genetics , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Knockout , Mice, Transgenic , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/biosynthesis , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/metabolism , Tissue Culture Techniques , Unfolded Protein Response/drug effects
7.
J Biol Chem ; 283(50): 34819-32, 2008 Dec 12.
Article in English | MEDLINE | ID: mdl-18936091

ABSTRACT

Endoplasmic reticulum (ER) stress induces INS-1 cell apoptosis by a pathway involving Ca(2+)-independent phospholipase A(2) (iPLA(2)beta)-mediated ceramide generation, but the mechanism by which iPLA(2)beta and ceramides contribute to apoptosis is not well understood. We report here that both caspase-12 and caspase-3 are activated in INS-1 cells following induction of ER stress with thapsigargin, but only caspase-3 cleavage is amplified in iPLA(2)beta overexpressing INS-1 cells (OE), relative to empty vector-transfected cells, and is suppressed by iPLA(2)beta inhibition. ER stress also led to the release of cytochrome c and Smac and, unexpectedly, their accumulation in the cytosol is amplified in OE cells. These findings raise the likelihood that iPLA(2)beta participates in ER stress-induced apoptosis by activating the intrinsic apoptotic pathway. Consistent with this possibility, we find that ER stress promotes iPLA(2)beta accumulation in the mitochondria, opening of mitochondrial permeability transition pore, and loss in mitochondrial membrane potential (Delta Psi) in INS-1 cells and that these changes are amplified in OE cells. ER stress also led to greater ceramide generation in ER and mitochondria fractions of OE cells. Exposure to ceramide alone induces loss in Delta Psi and apoptosis and these are suppressed by forskolin. ER stress-induced mitochondrial dysfunction and apoptosis are also inhibited by forskolin, as well as by inactivation of iPLA(2)beta or NSMase, suggesting that iPLA(2)beta-mediated generation of ceramides via sphingomyelin hydrolysis during ER stress affect the mitochondria. In support, inhibition of iPLA(2)beta or NSMase prevents cytochrome c release. Collectively, our findings indicate that the iPLA(2)beta-ceramide axis plays a critical role in activating the mitochondrial apoptotic pathway in insulin-secreting cells during ER stress.


Subject(s)
Apoptosis , Ceramides/metabolism , Endoplasmic Reticulum/metabolism , Group VI Phospholipases A2/metabolism , Insulin/metabolism , Mitochondria/metabolism , Animals , Caspase 12/metabolism , Caspase 3/metabolism , Cell Line , Group VI Phospholipases A2/chemistry , Group VI Phospholipases A2/physiology , Membrane Potentials , Models, Biological , Oxidative Stress , Rats , Sphingomyelins/metabolism
8.
Am J Physiol Endocrinol Metab ; 294(2): E217-29, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17895289

ABSTRACT

Studies with genetically modified insulinoma cells suggest that group VIA phospholipase A(2) (iPLA(2)beta) participates in amplifying glucose-induced insulin secretion. INS-1 insulinoma cells that overexpress iPLA(2)beta, for example, exhibit amplified insulin-secretory responses to glucose and cAMP-elevating agents. To determine whether similar effects occur in whole animals, we prepared transgenic (TG) mice in which the rat insulin 1 promoter (RIP) drives iPLA(2)beta overexpression, and two characterized TG mouse lines exhibit similar phenotypes. Their pancreatic islet iPLA(2)beta expression is increased severalfold, as reflected by quantitative PCR of iPLA(2)beta mRNA, immunoblotting of iPLA(2)beta protein, and iPLA(2)beta enzymatic activity. Immunofluorescence microscopic studies of pancreatic sections confirm iPLA(2)beta overexpression in RIP-iPLA(2)beta-TG islet beta-cells without obviously perturbed islet morphology. Male RIP-iPLA(2)beta-TG mice exhibit lower blood glucose and higher plasma insulin concentrations than wild-type (WT) mice when fasting and develop lower blood glucose levels in glucose tolerance tests, but WT and TG blood glucose levels do not differ in insulin tolerance tests. Islets from male RIP-iPLA(2)beta-TG mice exhibit greater amplification of glucose-induced insulin secretion by a cAMP-elevating agent than WT islets. In contrast, islets from male iPLA(2)beta-null mice exhibit blunted insulin secretion, and those mice have impaired glucose tolerance. Arachidonate incorporation into and the phospholipid composition of RIP-iPLA(2)beta-TG islets are normal, but they exhibit reduced Kv2.1 delayed rectifier current and prolonged glucose-induced action potentials and elevations of cytosolic Ca(2+) concentration that suggest a molecular mechanism for the physiological role of iPLA(2)beta to amplify insulin secretion.


Subject(s)
Blood Glucose/physiology , Group IV Phospholipases A2/biosynthesis , Homeostasis/physiology , Insulin/metabolism , Islets of Langerhans/metabolism , Phospholipids/metabolism , Animals , Arachidonic Acid/metabolism , Blood Glucose/metabolism , Blotting, Western , Calcium/physiology , Cell Line, Tumor , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Fasting/metabolism , Gene Expression Regulation, Enzymologic/physiology , Genotype , Glucose Tolerance Test , Group IV Phospholipases A2/genetics , Homeodomain Proteins/genetics , Insulin/blood , Insulin Secretion , Insulin-Secreting Cells/metabolism , Insulinoma/metabolism , Kv1.2 Potassium Channel/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Pancreatic Neoplasms/metabolism , Patch-Clamp Techniques , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Electrospray Ionization , Trans-Activators/genetics
9.
Biochemistry ; 46(35): 10170-85, 2007 Sep 04.
Article in English | MEDLINE | ID: mdl-17685585

ABSTRACT

Beta-cell mass is regulated by a balance between beta-cell growth and beta-cell death, due to apoptosis. We previously reported that apoptosis of INS-1 insulinoma cells due to thapsigargin-induced ER stress was suppressed by inhibition of the group VIA Ca2+-independent phospholipase A2 (iPLA2beta), associated with an increased level of ceramide generation, and that the effects of ER stress were amplified in INS-1 cells in which iPLA2beta was overexpressed (OE INS-1 cells). These findings suggested that iPLA2beta and ceramides participate in ER stress-induced INS-1 cell apoptosis. Here, we address this possibility and also the source of the ceramides by examining the effects of ER stress in empty vector (V)-transfected and iPLA2beta-OE INS-1 cells using apoptosis assays and immunoblotting, quantitative PCR, and mass spectrometry analyses. ER stress induced expression of ER stress factors GRP78 and CHOP, cleavage of apoptotic factor PARP, and apoptosis in V and OE INS-1 cells. Accumulation of ceramide during ER stress was not associated with changes in mRNA levels of serine palmitoyltransferase (SPT), the rate-limiting enzyme in de novo synthesis of ceramides, but both message and protein levels of neutral sphingomyelinase (NSMase), which hydrolyzes sphingomyelins to generate ceramides, were temporally increased in the INS-1 cells. The increases in the level of NSMase expression in the ER-stressed INS-1 cells were associated with corresponding temporal elevations in ER-associated iPLA2beta protein and catalytic activity. Pretreatment with BEL inactivated iPLA2beta and prevented induction of NSMase message and protein in ER-stressed INS-1 cells. Relative to that in V INS-1 cells, the effects of ER stress were accelerated and/or amplified in the OE INS-1 cells. However, inhibition of iPLA2beta or NSMase (chemically or with siRNA) suppressed induction of NSMase message, ceramide generation, sphingomyelin hydrolysis, and apoptosis in both V and OE INS-1 cells during ER stress. In contrast, inhibition of SPT did not suppress ceramide generation or apoptosis in either V or OE INS-1 cells. These findings indicate that iPLA2beta activation participates in ER stress-induced INS-1 cell apoptosis by promoting ceramide generation via NSMase-catalyzed hydrolysis of sphingomyelins, raising the possibility that this pathway contributes to beta-cell apoptosis due to ER stress.


Subject(s)
Apoptosis , Ceramides/metabolism , Group VI Phospholipases A2/metabolism , Insulinoma/enzymology , Pancreatic Neoplasms/enzymology , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelins/metabolism , Apoptosis/physiology , Cell Line, Tumor , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation, Enzymologic , Humans , Hydrolysis , Phosphodiesterase Inhibitors/pharmacology , RNA, Small Interfering , Retroviridae/genetics , Spectrometry, Mass, Electrospray Ionization , Sphingomyelin Phosphodiesterase/drug effects , Thapsigargin/pharmacology , Transfection
10.
J Biol Chem ; 282(37): 27100-27114, 2007 Sep 14.
Article in English | MEDLINE | ID: mdl-17627946

ABSTRACT

Mouse macrophages undergo ER stress and apoptosis upon free cholesterol loading (FCL). We recently generated iPLA(2)beta-null mice, and here we demonstrate that iPLA(2)beta-null macrophages have reduced sensitivity to FCL-induced apoptosis, although they and wild-type (WT) cells exhibit similar increases in the transcriptional regulator CHOP. iPLA(2)beta-null macrophages are also less sensitive to apoptosis induced by the sarcoplasmic reticulum Ca(2+)-ATPase inhibitor thapsigargin and the scavenger receptor A ligand fucoidan, and restoring iPLA(2)betaexpression with recombinant adenovirus increases apoptosis toward WT levels. WT and iPLA(2)beta-null macrophages incorporate [(3)H]arachidonic acid ([(3)H]AA]) into glycerophosphocholine lipids equally rapidly and exhibit identical zymosan-induced, cPLA(2)alpha-catalyzed [(3)H]AA release. In contrast, although WT macrophages exhibit robust [(3)H]AA release upon FCL, this is attenuated in iPLA(2)beta-null macrophages and increases toward WT levels upon restoring iPLA(2)beta expression. Recent reports indicate that iPLA(2)beta modulates mitochondrial cytochrome c release, and we find that thapsigargin and fucoidan induce mitochondrial phospholipid loss and cytochrome c release into WT macrophage cytosol and that these events are blunted in iPLA(2)beta-null cells. Immunoblotting studies indicate that iPLA(2)beta associates with mitochondria in macrophages subjected to ER stress. AA incorporation into glycerophosphocholine lipids is unimpaired in iPLA(2)beta-null macrophages upon electrospray ionization-tandem mass spectrometry analyses, and their complex lipid composition is similar to WT cells. These findings suggest that iPLA(2)beta participates in ER stress-induced macrophage apoptosis caused by FCL or thapsigargin but that deletion of iPLA(2)beta does not impair macrophage arachidonate incorporation or phospholipid composition.


Subject(s)
Apoptosis , Cholesterol/metabolism , Macrophages, Peritoneal/cytology , Phospholipases A/physiology , Phospholipids/analysis , Animals , Arachidonic Acid/metabolism , Cytochromes c/metabolism , Endoplasmic Reticulum/metabolism , Female , Group VI Phospholipases A2 , Macrophages, Peritoneal/chemistry , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mitochondria/chemistry , Phospholipases A/analysis , Phospholipases A/genetics , Phospholipases A2 , Polysaccharides/pharmacology , RNA, Messenger/analysis , Sphingolipids/analysis , Thapsigargin/pharmacology
11.
J Biol Chem ; 281(30): 20958-20973, 2006 Jul 28.
Article in English | MEDLINE | ID: mdl-16732058

ABSTRACT

Studies involving pharmacologic or molecular biologic manipulation of Group VIA phospholipase A(2) (iPLA(2)beta) activity in pancreatic islets and insulinoma cells suggest that iPLA(2)beta participates in insulin secretion. It has also been suggested that iPLA(2)beta is a housekeeping enzyme that regulates cell 2-lysophosphatidylcholine (LPC) levels and arachidonate incorporation into phosphatidylcholine (PC). We have generated iPLA(2)beta-null mice by homologous recombination and have reported that they exhibit reduced male fertility and defective motility of spermatozoa. Here we report that pancreatic islets from iPLA(2)beta-null mice have impaired insulin secretory responses to D-glucose and forskolin. Electrospray ionization mass spectrometric analyses indicate that the abundance of arachidonate-containing PC species of islets, brain, and other tissues from iPLA(2)beta-null mice is virtually identical to that of wild-type mice, and no iPLA(2)beta mRNA was observed in any tissue from iPLA(2)beta-null mice at any age. Despite the insulin secretory abnormalities of isolated islets, fasting and fed blood glucose concentrations of iPLA(2)beta-null and wild-type mice are essentially identical under normal circumstances, but iPLA(2)beta-null mice develop more severe hyperglycemia than wild-type mice after administration of multiple low doses of the beta-cell toxin streptozotocin, suggesting an impaired islet secretory reserve. A high fat diet also induces more severe glucose intolerance in iPLA(2)beta-null mice than in wild-type mice, but PLA(2)beta-null mice have greater responsiveness to exogenous insulin than do wild-type mice fed a high fat diet. These and previous findings thus indicate that iPLA(2)beta-null mice exhibit phenotypic abnormalities in pancreatic islets in addition to testes and macrophages.


Subject(s)
Blood Glucose/metabolism , Insulin/metabolism , Islets of Langerhans/cytology , Phospholipases A/genetics , Phospholipases A/physiology , Animals , Female , Homeostasis , Insulin Secretion , Macrophages/metabolism , Male , Mice , Phenotype , Phospholipases A2 , Phospholipids/chemistry , Phospholipids/metabolism , Spectrometry, Mass, Electrospray Ionization , Streptozocin/pharmacology , Testis/metabolism
12.
J Biol Chem ; 281(1): 187-98, 2006 Jan 06.
Article in English | MEDLINE | ID: mdl-16286468

ABSTRACT

Studies involving pharmacologic inhibition or transient reduction of Group VIA phospholipase A2 (iPLA2beta) expression have suggested that it is a housekeeping enzyme that regulates cell 2-lysophosphatidylcholine (LPC) levels, rates of arachidonate incorporation into phospholipids, and degradation of excess phosphatidylcholine (PC). In insulin-secreting islet beta-cells and some other cells, in contrast, iPLA2beta signaling functions have been proposed. Using retroviral vectors, we prepared clonal INS-1 beta-cell lines in which iPLA2beta expression is stably suppressed by small interfering RNA. Two such iPLA2beta knockdown (iPLA2beta-KD) cell lines express less than 20% of the iPLA2beta of control INS-1 cell lines. The iPLA2beta-KD INS-1 cells exhibit impaired insulin secretory responses and reduced proliferation rates. Electrospray ionization mass spectrometric analyses of PC and LPC species that accumulate in INS-1 cells cultured with arachidonic acid suggest that 18:0/20:4-glycerophosphocholine (GPC) synthesis involves sn-2 remodeling to yield 16:0/20:4-GPC and then sn-1 remodeling via a 1-lyso/20:4-GPC intermediate. Electrospray ionization mass spectrometric analyses also indicate that the PC and LPC content and composition of iPLA2beta-KD and control INS-1 cells are nearly identical, as are the rates of arachidonate incorporation into PC and the composition and remodeling of other phospholipid classes. These findings indicate that iPLA2beta plays signaling or effector roles in beta-cell secretion and proliferation but that stable suppression of its expression does not affect beta-cell GPC lipid content or composition even under conditions in which LPC is being actively consumed by conversion to PC. This calls into question the generality of proposed housekeeping functions for iPLA2beta in PC homeostasis and remodeling.


Subject(s)
Insulin-Secreting Cells/enzymology , Insulin/metabolism , Phospholipases A/genetics , Phospholipases A/metabolism , Animals , Arachidonic Acid/pharmacokinetics , Cell Division/physiology , Cell Line, Tumor , Gene Expression Regulation, Enzymologic , Glycerophosphates/metabolism , Group IV Phospholipases A2 , Inositol Phosphates/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Insulinoma , Pancreatic Neoplasms , Phosphatidylethanolamines/metabolism , Phospholipases A2 , Phospholipids/metabolism , RNA, Small Interfering , Rats , Spectrometry, Mass, Electrospray Ionization , Transfection , Tritium
13.
Biochemistry ; 43(4): 918-30, 2004 Feb 03.
Article in English | MEDLINE | ID: mdl-14744135

ABSTRACT

The death of insulin-secreting beta-cells that causes type I diabetes mellitus (DM) occurs in part by apoptosis, and apoptosis also contributes to progressive beta-cell dysfunction in type II DM. Recent reports indicate that ER stress-induced apoptosis contributes to beta-cell loss in diabetes. Agents that deplete ER calcium levels induce beta-cell apoptosis by a process that is independent of increases in [Ca(2+)](i). Here we report that the SERCA inhibitor thapsigargin induces apoptosis in INS-1 insulinoma cells and that this is inhibited by a bromoenol lactone (BEL) inhibitor of group VIA calcium-independent phospholipase A(2) (iPLA(2)beta). Overexpression of iPLA(2)beta amplifies thapsigargin-induced apoptosis of INS-1 cells, and this is also suppressed by BEL. The magnitude of thapsigargin-induced INS-1 cell apoptosis correlates with the level of iPLA(2)beta expression in various cell lines, and apoptosis is associated with stimulation of iPLA(2)beta activity, perinuclear accumulation of iPLA(2)beta protein and activity, and caspase-3-catalyzed cleavage of full-length 84 kDa iPLA(2)beta to a 62 kDa product that associates with nuclei. Thapsigargin also induces ceramide accumulation in INS-1 cells, and this response is amplified in cells that overexpress iPLA(2)beta. These findings indicate that iPLA(2)beta participates in ER stress-induced apoptosis, a pathway that promotes beta-cell death in diabetes.


Subject(s)
Apoptosis , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/physiology , Insulin/metabolism , Phospholipases A/antagonists & inhibitors , Phospholipases A/biosynthesis , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Ceramides/metabolism , Dimethyl Sulfoxide/pharmacology , Endoplasmic Reticulum/enzymology , Fluorescent Antibody Technique, Indirect , Green Fluorescent Proteins , Group IV Phospholipases A2 , Group VI Phospholipases A2 , Immunoblotting , Insulin Secretion , Isoenzymes/antagonists & inhibitors , Isoenzymes/biosynthesis , Isoenzymes/genetics , Luminescent Proteins/biosynthesis , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Molecular Weight , Naphthalenes/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Phospholipases A/genetics , Phospholipases A2 , Pyrones/pharmacology , Rats , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Subcellular Fractions/drug effects , Subcellular Fractions/enzymology , Thapsigargin/pharmacology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL