Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Case Rep Emerg Med ; 2018: 5740509, 2018.
Article in English | MEDLINE | ID: mdl-29862090

ABSTRACT

Reported is a case of a 39-year-old Caucasian man who presented to the emergency department with sudden onset bilateral lower extremity paralysis after consuming a large amount of carbohydrates and alcohol. A CT, MRI, and lumbar puncture were performed with negative results; lab results showed hyperthyroidism and hypokalemia. The patient was diagnosed with thyrotoxic periodic paralysis. In a patient presenting with sudden onset paralysis and hypokalemia, the emergency physician should include thyrotoxic periodic paralysis in the differential diagnosis and focus on treating and working up the hypokalemia instead of the paralysis.

2.
Arterioscler Thromb Vasc Biol ; 34(4): 768-78, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24482376

ABSTRACT

OBJECTIVE: We previously demonstrated that nicotinamide adenine dinucleotide phosphate oxidase 4 (Nox4) mediates increased monocyte priming and chemotaxis under conditions of diabetic metabolic stress, and emerging data indicate that group VIA phospholipase A2 (iPLA2ß) also participates in regulating monocyte chemotaxis. Here, we examined relationships between iPLA2ß expression and Nox4 action in mouse peritoneal macrophages subjected to diabetic metabolic stress. APPROACH AND RESULTS: Increased iPLA2ß expression and activity were observed in macrophages from low-density lipoprotein receptor knockout mice that were fed a high-fat diet, and this was associated with time-dependent increases in atherosclerotic lesion size and macrophage content. Incubating macrophages with 30 mmol/L D-glucose, 100 µg/mL low-density lipoprotein, or both (D-glucose+low-density lipoprotein) induced a robust increase in iPLA2ß expression and activity and in cell migration in response to monocyte chemoattractant protein-1. The increases in iPLA2ß activity and cell migration were prevented by a bromoenol lactone iPLA2ß suicide inhibitor or an iPLA2ß antisense oligonucleotide. Incubating macrophages under conditions that mimic diabetic metabolic stress ex vivo resulted in increased Nox4 expression and activity and hydrogen peroxide generation compared with controls. Bromoenol lactone prevented those effects without affecting Nox2 expression. Nox4 inhibition eliminated diabetic metabolic stress-induced acceleration of macrophage migration. Lysophosphatidic acid restored Nox4 expression, hydrogen peroxide generation, and migration to bromoenol lactone-treated cells, and a lysophosphatidic acid receptor antagonist abrogated iPLA2ß-mediated increases in Nox4 expression. CONCLUSIONS: Taken together, these observations identify iPLA2ß and lysophosphatidic acid derived from its action as critical in regulating macrophage Nox4 activity and migration in the diabetic state in vivo and under similar conditions ex vivo.


Subject(s)
Atherosclerosis/enzymology , Cell Movement , Diabetes Mellitus/enzymology , Group VI Phospholipases A2/metabolism , Macrophages, Peritoneal/enzymology , NADPH Oxidases/metabolism , Signal Transduction , Animals , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Cells, Cultured , Diabetes Mellitus/genetics , Diabetes Mellitus/immunology , Diabetes Mellitus/pathology , Diet, High-Fat , Disease Models, Animal , Disease Progression , Enzyme Inhibitors/pharmacology , Group VI Phospholipases A2/genetics , Hydrogen Peroxide/metabolism , Lysophospholipids/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 4 , NADPH Oxidases/genetics , Oligonucleotides, Antisense/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics , Stress, Physiological , Time Factors , Transfection , Up-Regulation
3.
Endocr Pract ; 20(3): e42-6, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24246337

ABSTRACT

OBJECTIVE: To demonstrate that xanthogranuloma is a rare cause of idiopathic central diabetes insipidus in the early phase of the disease and that it presents as a suprasellar mass at a later stage. In addition, we emphasize the importance of identifying the cause of idiopathic central diabetes insipidus and review the literature concerning endocrine disturbance in central xanthogranuloma. METHODS: Review of recently published case reports of central xanthogranuloma with endocrine disorders. The case of a 35-year-old man who presented with a very large suprasellar mass is also reported. The patient was diagnosed with idiopathic central diabetes insipidus 20 years ago with normal brain magnetic resonance imaging. RESULTS: Most cases of this disease present as supra- or parasellar masses with endocrine involvement, the most common of which (in approximately 75% of patients) is sex hormone deficiency. Diabetes insipidus was found in 65% of patients. CONCLUSION: Xanthogranuloma should be in the differential diagnosis of idiopathic central diabetes insipidus and sellar and parasellar masses. A detailed skin examination is very important in making the diagnosis of central diabetes insipidus.

4.
Oxid Med Cell Longev ; 2012: 989372, 2012.
Article in English | MEDLINE | ID: mdl-23213352

ABSTRACT

Group VIB Phospholipase A(2) (iPLA(2)γ) is distributed in membranous organelles in which ß-oxidation occurs, that is, mitochondria and peroxisomes, and is expressed by insulin-secreting pancreatic islet ß-cells and INS-1 insulinoma cells, which can be injured by inflammatory cytokines, for example, IL-1ß and IFN-γ, and by oxidants, for example, streptozotocin (STZ) or t-butyl-hydroperoxide (TBHP), via processes pertinent to mechanisms of ß-cell loss in types 1 and 2 diabetes mellitus. We find that incubating INS-1 cells with IL-1ß and IFN-γ, with STZ, or with TBHP causes increased expression of iPLA(2)γ mRNA and protein. We prepared INS-1 knockdown (KD) cell lines with reduced iPLA(2)γ expression, and they proliferate more slowly than control INS-1 cells and undergo increased membrane peroxidation in response to cytokines or oxidants. Accumulation of oxidized phospholipid molecular species in STZ-treated INS-1 cells was demonstrated by LC/MS/MS scanning, and the levels in iPLA(2)γ-KD cells exceeded those in control cells. iPLA(2)γ-KD INS-1 cells also exhibited higher levels of apoptosis than control cells when incubated with STZ or with IL-1ß and IFN-γ. These findings suggest that iPLA(2)γ promotes ß-cell proliferation and that its expression is increased during inflammation or oxidative stress as a mechanism to mitigate membrane injury that may enhance ß-cell survival.


Subject(s)
Apoptosis/drug effects , Cytokines/pharmacology , Group VI Phospholipases A2/metabolism , Insulinoma/enzymology , Insulinoma/pathology , Lipid Peroxidation/drug effects , Oxidants/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatography, High Pressure Liquid , Gene Knockdown Techniques , Inflammation Mediators/pharmacology , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Lipids/chemistry , Oxidation-Reduction/drug effects , Rats , Spectrometry, Mass, Electrospray Ionization , Streptozocin/pharmacology , tert-Butylhydroperoxide/pharmacology
6.
J Biol Chem ; 287(8): 5528-41, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22194610

ABSTRACT

Group VIA phospholipase A(2) (iPLA(2)ß) in pancreatic islet ß-cells participates in glucose-stimulated insulin secretion and sarco(endo)plasmic reticulum ATPase (SERCA) inhibitor-induced apoptosis, and both are attenuated by pharmacologic or genetic reductions in iPLA(2)ß activity and amplified by iPLA(2)ß overexpression. While exploring signaling events that occur downstream of iPLA(2)ß activation, we found that p38 MAPK is activated by phosphorylation in INS-1 insulinoma cells and mouse pancreatic islets, that this increases with iPLA(2)ß expression level, and that it is stimulated by the iPLA(2)ß reaction product arachidonic acid. The insulin secretagogue D-glucose also stimulates ß-cell p38 MAPK phosphorylation, and this is prevented by the iPLA(2)ß inhibitor bromoenol lactone. Insulin secretion induced by d-glucose and forskolin is amplified by overexpressing iPLA(2)ß in INS-1 cells and in mouse islets, and the p38 MAPK inhibitor PD169316 prevents both responses. The SERCA inhibitor thapsigargin also stimulates phosphorylation of both ß-cell MAPK kinase isoforms and p38 MAPK, and bromoenol lactone prevents both events. Others have reported that iPLA(2)ß products activate Rho family G-proteins that promote MAPK kinase activation via a mechanism inhibited by Clostridium difficile toxin B, which we find to inhibit thapsigargin-induced ß-cell p38 MAPK phosphorylation. Thapsigargin-induced ß-cell apoptosis and ceramide generation are also prevented by the p38 MAPK inhibitor PD169316. These observations indicate that p38 MAPK is activated downstream of iPLA(2)ß in ß-cells incubated with insulin secretagogues or thapsigargin, that this requires prior iPLA(2)ß activation, and that p38 MAPK is involved in the ß-cell functional responses of insulin secretion and apoptosis in which iPLA(2)ß participates.


Subject(s)
Group VI Phospholipases A2/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Arachidonic Acid/pharmacology , Cell Line, Tumor , Ceramides/metabolism , Dose-Response Relationship, Drug , Endoplasmic Reticulum Stress/drug effects , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Glucose/pharmacology , Group VI Phospholipases A2/antagonists & inhibitors , Imidazoles/pharmacology , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/enzymology , Male , Mice , Naphthalenes/pharmacology , Phosphorylation/drug effects , Pyrones/pharmacology , Rats , Signal Transduction/drug effects , Thapsigargin/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , rho GTP-Binding Proteins/metabolism
7.
Endocr Pract ; 18(2): e26-31, 2012.
Article in English | MEDLINE | ID: mdl-22138080

ABSTRACT

OBJECTIVES: To indicate cardiogenic shock as a very rare but serious clinical consequence of untreated panhypopituitarism attributable to Sheehan syndrome; to emphasize the importance of eliciting a detailed endocrine and obstetric history in women presenting with idiopathic heart failure; to highlight the diagnostic shortcomings of screening for thyroid dysfunction solely with thyroid-stimulating hormone determinations; and to report the reversibility of severe heart failure induced by long-term pituitary insufficiency. METHODS: Described is a case report of a 35-year-old woman who presented with severe congestive heart failure, hypotension, and confusion. Her 2-dimensional echocardiogram revealed appreciable systolic and diastolic dysfunction. In screening for possible endocrine causes of heart failure, a normal thyroid-stimulating hormone level of 0.72 mIU/L (reference range, 0.35 to 5.5) was unremarkable; however, a profoundly low free thyroxine level of 0.12 ng/dL (reference range, 0.9 to 1.8) led clinicians to pursue a work-up of central hypothyroidism. RESULTS: Endocrine testing confirmed the presence of panhypopituitarism and adrenal insufficiency. Magnetic resonance imaging of the brain revealed empty sella syndrome. Further questioning of the patient revealed a history of extensive postpartum bleeding 15 years earlier, failure to lactate, and secondary amenorrhea--all consistent with undiagnosed Sheehan syndrome. In the hospital, the patient was treated with intravenously administered corticosteroids and levothyroxine. Her mental status and symptomatic heart failure improved dramatically. After 9 months of oral levothyroxine and glucocorticoid therapy, the patient remained asymptomatic, and repeated echocardiography indicated completely normalized cardiac function. CONCLUSION: Severe heart failure and cardiogenic shock can be a very rare (but fortunately reversible) complication of long-standing panhypopituitarism resulting from undiagnosed Sheehan syndrome.


Subject(s)
Delayed Diagnosis/adverse effects , Heart Failure/etiology , Hormone Replacement Therapy , Hypopituitarism/drug therapy , Hypopituitarism/physiopathology , Shock, Cardiogenic/etiology , Adult , Confusion/etiology , Confusion/prevention & control , Drug Therapy, Combination , Female , Glucocorticoids/therapeutic use , Heart Failure/physiopathology , Heart Failure/prevention & control , Humans , Hydrocortisone/therapeutic use , Hypopituitarism/diagnosis , Hypopituitarism/etiology , Hypotension/etiology , Hypotension/prevention & control , Prednisone/therapeutic use , Severity of Illness Index , Shock, Cardiogenic/prevention & control , Thyroxine/therapeutic use , Treatment Outcome
8.
Biochim Biophys Acta ; 1801(5): 547-58, 2010 May.
Article in English | MEDLINE | ID: mdl-20132906

ABSTRACT

Over the past decade, important roles for the 84-88kDa Group VIA Ca(2+)-independent phospholipase A(2) (iPLA(2)beta) in various organs have been described. We demonstrated that iPLA(2)beta participates in insulin secretion, insulinoma cells and native pancreatic islets express full-length and truncated isoforms of iPLA(2)beta, and certain stimuli promote perinuclear localization of iPLA(2)beta. To gain a better understanding of its mobilization, iPLA(2)beta was expressed in INS-1 cells as a fusion protein with EGFP, enabling detection of subcellular localization of iPLA(2)beta by monitoring EGFP fluorescence. Cells stably-transfected with fusion protein expressed nearly 5-fold higher catalytic iPLA(2)beta activity than control cells transfected with EGFP cDNA alone, indicating that co-expression of EGFP does not interfere with manifestation of iPLA(2)beta activity. Dual fluorescence monitoring of EGFP and organelle Trackers combined with immunoblotting analyses revealed expression of truncated iPLA(2)beta isoforms in separate subcellular organelles. Exposure to secretagogues and induction of ER stress are known to activate iPLA(2)beta in beta-cells and we find here that these stimuli promote differential localization of iPLA(2)beta in subcellular organelles. Further, mass spectrometric analyses identified iPLA(2)beta variants from which N-terminal residues were removed. Collectively, these findings provide evidence for endogenous proteolytic processing of iPLA(2)beta and redistribution of iPLA(2)beta variants in subcellular compartments. It might be proposed that in vivo processing of iPLA(2)beta facilitates its participation in multiple biological processes.


Subject(s)
Group VI Phospholipases A2/metabolism , Organelles/metabolism , Protein Isoforms/metabolism , Recombinant Fusion Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Endoplasmic Reticulum/metabolism , Group VI Phospholipases A2/genetics , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Mass Spectrometry , Mitochondria/metabolism , Molecular Sequence Data , Oxidative Stress , Protein Isoforms/genetics , Rats , Recombinant Fusion Proteins/genetics , Subcellular Fractions/metabolism
9.
Am J Physiol Endocrinol Metab ; 298(6): E1097-114, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20179248

ABSTRACT

Phospholipases A(2) (PLA(2)) play important roles in metabolic processes, and the Group VI PLA(2) family is comprised of intracellular enzymes that do not require Ca(2+) for catalysis. Mice deficient in Group VIA PLA(2) (iPLA(2)beta) develop more severe glucose intolerance than wild-type (WT) mice in response to dietary stress. Group VIB PLA(2) (iPLA(2)gamma) is a related enzyme distributed in membranous organelles, including mitochondria, and iPLA(2)gamma knockout (KO) mice exhibit altered mitochondrial morphology and function. We have compared metabolic responses of iPLA(2)gamma-KO and WT mice fed a Western diet (WD) with a high fat content. We find that KO mice are resistant to WD-induced increases in body weight and adiposity and in blood levels of cholesterol, glucose, and insulin, even though WT and KO mice exhibit similar food consumption and dietary fat digestion and absorption. KO mice are also relatively resistant to WD-induced insulin resistance, glucose intolerance, and altered patterns of fat vs. carbohydrate fuel utilization. KO skeletal muscle exhibits impaired mitochondrial beta-oxidation of fatty acids, as reflected by accumulation of larger amounts of long-chain acylcarnitine (LCAC) species in KO muscle and liver compared with WT in response to WD feeding. This is associated with increased urinary excretion of LCAC and much reduced deposition of triacylglycerols in liver by WD-fed KO compared with WT mice. The iPLA(2)gamma-deficient genotype thus results in a phenotype characterized by impaired mitochondrial oxidation of fatty acids and relative resistance to the metabolic abnormalities induced by WD.


Subject(s)
Group IV Phospholipases A2/deficiency , Obesity/enzymology , 3-Hydroxybutyric Acid/blood , Animals , Blood Glucose/metabolism , Body Composition/physiology , Cholesterol/blood , Dietary Fats/metabolism , Fatty Acids, Nonesterified/blood , Feces/chemistry , Female , Glycerol/blood , Group IV Phospholipases A2/metabolism , Insulin/blood , Insulin Resistance/physiology , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Obesity/blood , Random Allocation , Specific Pathogen-Free Organisms , Triglycerides/blood
10.
J Innate Immun ; 1(6): 607-17, 2009.
Article in English | MEDLINE | ID: mdl-20160900

ABSTRACT

Calcium-independent phospholipase A2 (iPLA2) has been suggested to play an important role in the activation of caspase-1 induced by lipopolysaccharides (LPS). Here, we used pharmacological and genetic approaches to study the role of iPLA 2 in the activation of caspase-1. Bromoenol lactone (BEL), an inhibitor that was originally used to support a role for iPLA2 in the secretion of IL-1 beta, prevented caspase-1 activation induced by LPS and ATP as described, and also activation triggered by Salmonella infection and cytosolic flagellin, which rely on the Nlrc4 inflammasome. Analysis of BEL enantiomers showed that the S-BEL form was more effective than R-BEL in inhibiting the inflammasome, suggesting a role for iPLA2 . However, caspase-1 activation and IL-1 beta secretion and their inhibition by BEL were unimpaired in macrophages deficient in iPLA2 beta. BEL was originally identified as an inhibitor of serine proteases. Consistent with the latter, the serine proteases inhibitors TPCK, TLCK and AAF-cmk prevented the activation of the Nlrc4 and Nlrp3 inflammasomes while pan-cathepsin inhibitors were ineffective. These results indicate that iPLA2 beta is not critical for caspase-1 activation as currently proposed. Instead, the results suggest that serine protease(s) targeted by BEL may play a critical role in the activation of the inflammasome triggered by microbial stimuli.


Subject(s)
Enzyme Activation/physiology , Group IV Phospholipases A2/metabolism , Macrophages/metabolism , Naphthalenes/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Phospholipases A2, Calcium-Independent/metabolism , Pyrones/pharmacology , Animals , Apoptosis Regulatory Proteins/immunology , Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/immunology , Calcium-Binding Proteins/metabolism , Carrier Proteins/immunology , Carrier Proteins/metabolism , Caspase 1/immunology , Caspase 1/metabolism , Enzyme Activation/drug effects , Group IV Phospholipases A2/immunology , Immunoblotting , Inflammation/enzymology , Inflammation/immunology , Macrophages/immunology , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Phospholipases A2, Calcium-Independent/immunology , Stereoisomerism
11.
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
12.
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
13.
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
14.
J Biol Chem ; 282(10): 7442-9, 2007 Mar 09.
Article in English | MEDLINE | ID: mdl-17197450

ABSTRACT

Glucose stimulates both insulin secretion and hydrolysis of arachidonic acid (AA) esterified in membrane phospholipids of pancreatic islet beta-cells, and these processes are amplified by muscarinic agonists. Here we demonstrate that nonesterified AA regulates the biophysical activity of the pancreatic islet beta-cell-delayed rectifier channel, Kv2.1. Recordings of Kv2.1 currents from INS-1 insulinoma cells incubated with AA (5 mum) and subjected to graded degrees of depolarization exhibit a significantly shorter time-to-peak current interval than do control cells. AA causes a rapid decay and reduced peak conductance of delayed rectifier currents from INS-1 cells and from primary beta-cells isolated from mouse, rat, and human pancreatic islets. Stimulating mouse islets with AA results in a significant increase in the frequency of glucose-induced [Ca(2+)] oscillations, which is an expected effect of Kv2.1 channel blockade. Stimulation with concentrations of glucose and carbachol that accelerate hydrolysis of endogenous AA from islet phosphoplipids also results in accelerated Kv2.1 inactivation and a shorter time-to-peak current interval. Group VIA phospholipase A(2) (iPLA(2)beta) hydrolyzes beta-cell membrane phospholipids to release nonesterified fatty acids, including AA, and inhibiting iPLA(2)beta prevents the muscarinic agonist-induced accelerated Kv2.1 inactivation. Furthermore, glucose and carbachol do not significantly affect Kv2.1 inactivation in beta-cells from iPLA(2)beta(-/-) mice. Stably transfected INS-1 cells that overexpress iPLA(2)beta hydrolyze phospholipids more rapidly than control INS-1 cells and also exhibit an increase in the inactivation rate of the delayed rectifier currents. These results suggest that Kv2.1 currents could be dynamically modulated in the pancreatic islet beta-cell by phospholipase-catalyzed hydrolysis of membrane phospholipids to yield non-esterified fatty acids, such as AA, that facilitate Ca(2+) entry and insulin secretion.


Subject(s)
Arachidonic Acid/pharmacology , Islets of Langerhans/drug effects , Shab Potassium Channels/drug effects , Animals , Calcium/metabolism , Carbachol/pharmacology , Cell Line , Humans , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Phospholipases A/physiology , Rats , Shab Potassium Channels/physiology
15.
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
16.
Biochemistry ; 45(20): 6392-406, 2006 May 23.
Article in English | MEDLINE | ID: mdl-16700550

ABSTRACT

Group VIA phospholipase A(2) (iPLA(2)beta) is expressed in phagocytes, vascular cells, pancreatic islet beta-cells, neurons, and other cells and plays roles in transcriptional regulation, cell proliferation, apoptosis, secretion, and other events. A bromoenol lactone (BEL) suicide substrate used to study iPLA(2)beta functions inactivates iPLA(2)beta by alkylating Cys thiols. Because thiol redox reactions are important in signaling and some cells that express iPLA(2)beta produce biological oxidants, iPLA(2)beta might be subject to redox regulation. We report that biological concentrations of H(2)O(2), NO, and HOCl inactivate iPLA(2)beta, and this can be partially reversed by dithiothreitol (DTT). Oxidant-treated iPLA(2)beta modifications were studied by LC-MS/MS analyses of tryptic digests and included DTT-reversible events, e.g., formation of disulfide bonds and sulfenic acids, and others not so reversed, e.g., formation of sulfonic acids, Trp oxides, and Met sulfoxides. W(460) oxidation could cause irreversible inactivation because it is near the lipase consensus sequence ((463)GTSTG(467)), and site-directed mutagenesis of W(460) yields active mutant enzymes that exhibit no DTT-irreversible oxidative inactivation. Cys651-sulfenic acid formation could be one DTT-reversible inactivation event because Cys651 modification correlates closely with activity loss and its mutagenesis reduces sensitivity to inhibition. Intermolecular disulfide bond formation might also cause reversible inactivation because oxidant-treated iPLA(2)beta contains DTT-reducible oligomers, and oligomerization occurs with time- and temperature-dependent iPLA(2)beta inactivation that is attenuated by DTT or ATP. Subjecting insulinoma cells to oxidative stress induces iPLA(2)beta oligomerization, loss of activity, and subcellular redistribution and reduces the rate of release of arachidonate from phospholipids. These findings raise the possibility that redox reactions affect iPLA(2)beta functions.


Subject(s)
Oxidants/pharmacology , Phospholipases A/chemistry , Phospholipases A/metabolism , Amino Acid Sequence , Animals , Catalysis/drug effects , Cells, Cultured , Chromatography, Liquid , Cysteine/metabolism , Dithiothreitol/metabolism , Dithiothreitol/pharmacology , Group VI Phospholipases A2 , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Hypochlorous Acid/metabolism , Hypochlorous Acid/pharmacology , Methionine/metabolism , Molecular Sequence Data , Nitric Oxide/metabolism , Nitric Oxide/pharmacology , Oxidants/metabolism , Oxidation-Reduction , Oxidative Stress , Peptide Hydrolases/metabolism , Phospholipases A/genetics , Phospholipases A2 , Spectrometry, Mass, Electrospray Ionization , Spodoptera/cytology , Temperature , Time Factors , Tryptophan/metabolism
17.
Biochemistry ; 45(3): 1061-73, 2006 Jan 24.
Article in English | MEDLINE | ID: mdl-16411783

ABSTRACT

Phospholipases A2 (PLA2) comprise a superfamily of enzymes that hydrolyze phospholipids to a free fatty acid, e.g., arachidonate, and a 2-lysophospholipid. Dissecting their individual functions has relied in large part on pharmacological inhibitors that discriminate among PLA2. Group VIA PLA2 (iPLA2beta) has a GTSTG serine lipase consensus sequence, and studies with a bromoenol lactone (BEL) suicide substrate inhibitor have been taken to suggest that iPLA2beta participates in a wide variety of biological processes. Such conclusions presume inhibitor specificity. Inhibition by BEL requires its hydrolysis by and results in uncharacterized covalent modification(s) of iPLA2beta. We performed mass spectrometric analyses of proteolytic digests of BEL-treated iPLA2beta to identify modifications associated with loss of activity. The GTSTG active site and large flanking regions of sequence are not modified by BEL treatment, but most iPLA2beta Cys residues are alkylated at various BEL concentrations to form a thioether linkage to a BEL keto acid hydrolysis product. Synthetic Cys-containing peptides are alkylated when incubated with iPLA2beta and BEL, which reflects iPLA2beta-catalyzed BEL hydrolysis to a diffusible bromomethyl keto acid product that reacts with distant thiols. The BEL concentration dependence of Cys651 alkylation closely parallels that of loss of iPLA2beta activity. No amino acid residues other than Cys were found to be modified, suggesting that Cys alkylation is the covalent modification of iPLA2beta responsible for loss of activity, and the alkylating species appears to be a diffusible hydrolysis product of BEL rather than a tethered acyl-enzyme intermediate.


Subject(s)
Cysteine/chemistry , Enzyme Inhibitors/pharmacology , Hydrocarbons, Brominated/chemistry , Keto Acids/chemistry , Naphthalenes/metabolism , Phospholipases A/antagonists & inhibitors , Pyrones/metabolism , Sulfhydryl Compounds/metabolism , Amino Acid Sequence , Baculoviridae/genetics , Cells, Cultured , Diffusion , Enzyme Inhibitors/chemistry , Group II Phospholipases A2 , Models, Molecular , Molecular Sequence Data , Phospholipases A/chemistry , Phospholipases A/genetics , Phospholipases A/metabolism , Phospholipases A2 , Recombinant Proteins/metabolism , Substrate Specificity
18.
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
19.
J Biol Chem ; 280(8): 6840-9, 2005 Feb 25.
Article in English | MEDLINE | ID: mdl-15576376

ABSTRACT

Insulin-secreting pancreatic islet beta-cells express a Group VIA Ca(2+)-independent phospholipase A(2) (iPLA(2)beta) that contains a calmodulin binding site and protein interaction domains. We identified Ca(2+)/calmodulin-dependent protein kinase IIbeta (CaMKIIbeta) as a potential iPLA(2)beta-interacting protein by yeast two-hybrid screening of a cDNA library using iPLA(2)beta cDNA as bait. Cloning CaMKIIbeta cDNA from a rat islet library revealed that one dominant CaMKIIbeta isoform mRNA is expressed by adult islets and is not observed in brain or neonatal islets and that there is high conservation of the isoform expressed by rat and human beta-cells. Binary two-hybrid assays using DNA encoding this isoform as bait and iPLA(2)beta DNA as prey confirmed interaction of the enzymes, as did assays with CaMKIIbeta as prey and iPLA(2)beta bait. His-tagged CaMKIIbeta immobilized on metal affinity matrices bound iPLA(2)beta, and this did not require exogenous calmodulin and was not prevented by a calmodulin antagonist or the Ca(2+) chelator EGTA. Activities of both enzymes increased upon their association, and iPLA(2)beta reaction products reduced CaMKIIbeta activity. Both the iPLA(2)beta inhibitor bromoenol lactone and the CaMKIIbeta inhibitor KN93 reduced arachidonate release from INS-1 insulinoma cells, and both inhibit insulin secretion. CaMKIIbeta and iPLA(2)beta can be coimmunoprecipitated from INS-1 cells, and forskolin, which amplifies glucose-induced insulin secretion, increases the abundance of the immunoprecipitatable complex. These findings suggest that iPLA(2)beta and CaMKIIbeta form a signaling complex in beta-cells, consistent with reports that both enzymes participate in insulin secretion and that their expression is coinduced upon differentiation of pancreatic progenitor to endocrine progenitor cells.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Islets of Langerhans/enzymology , Phospholipases A/physiology , Signal Transduction , Animals , Arachidonic Acid/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calmodulin/pharmacology , Cell Line , Enzyme Inhibitors/pharmacology , Group VI Phospholipases A2 , Insecta , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Phospholipases A/metabolism , Phospholipases A2 , Protein Binding , RNA, Messenger/analysis , Rats , Tissue Distribution , Two-Hybrid System Techniques
20.
J Am Soc Mass Spectrom ; 15(12): 1780-93, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15589755

ABSTRACT

Dysregulation of proteolytic processing of the amyloid precursor protein (APP) contributes to the pathogenesis of Alzheimer's Disease, and the Group VIA phospholipase A(2) (iPLA(2)beta) is the dominant PLA(2) enzyme in the central nervous system and is subject to regulatory proteolytic processing. We have identified novel N-terminal variants of iPLA(2)beta and previously unrecognized proteolysis sites in APP constructs with a C-terminal 6-myc tag by automated identification of signature peptides in LC/MS/MS analyses of proteolytic digests. We have developed a Signature-Discovery (SD) program to characterize protein isoforms by identifying signature peptides that arise from proteolytic processing in vivo. This program analyzes MS/MS data from LC analyses of proteolytic digests of protein mixtures that can include incompletely resolved components in biological samples. This reduces requirements for purification and thereby minimizes artifactual modifications during sample processing. A new algorithm to generate the theoretical signature peptide set and to calculate similarity scores between predicted and observed mass spectra has been tested and optimized with model proteins. The program has been applied to the identification of variants of proteins of biological interest, including APP cleavage products and iPLA(2)beta, and such applications demonstrate the utility of this approach.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Peptide Hydrolases/chemistry , Peptides/analysis , Phospholipases A/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Spodoptera , Algorithms , Amino Acid Sequence , Animals , Cells, Cultured , Humans , Isoenzymes , Molecular Sequence Data , Phospholipases A2
SELECTION OF CITATIONS
SEARCH DETAIL
...