Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 289(40): 27410-7, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25122761

ABSTRACT

Group X secretory phospholipase A2 (GX sPLA2) potently hydrolyzes membrane phospholipids to release arachidonic acid (AA). While AA is an activator of glucose-stimulated insulin secretion (GSIS), its metabolite prostaglandin E2 (PGE2) is a known inhibitor. In this study, we determined that GX sPLA2 is expressed in insulin-producing cells of mouse pancreatic islets and investigated its role in beta cell function. GSIS was measured in vivo in wild-type (WT) and GX sPLA2-deficient (GX KO) mice and ex vivo using pancreatic islets isolated from WT and GX KO mice. GSIS was also assessed in vitro using mouse MIN6 pancreatic beta cells with or without GX sPLA2 overexpression or exogenous addition. GSIS was significantly higher in islets isolated from GX KO mice compared with islets from WT mice. Conversely, GSIS was lower in MIN6 cells overexpressing GX sPLA2 (MIN6-GX) compared with control (MIN6-C) cells. PGE2 production was significantly higher in MIN6-GX cells compared with MIN6-C cells and this was associated with significantly reduced cellular cAMP. The effect of GX sPLA2 on GSIS was abolished when cells were treated with NS398 (a COX-2 inhibitor) or L-798,106 (a PGE2-EP3 receptor antagonist). Consistent with enhanced beta cell function, GX KO mice showed significantly increased plasma insulin levels following glucose challenge and were protected from age-related reductions in GSIS and glucose tolerance compared with WT mice. We conclude that GX sPLA2 plays a previously unrecognized role in negatively regulating pancreatic insulin secretion by augmenting COX-2-dependent PGE2 production.


Subject(s)
Cyclooxygenase 2/metabolism , Group X Phospholipases A2/metabolism , Insulin-Secreting Cells/enzymology , Insulin/metabolism , Animals , Cyclooxygenase 2/genetics , Dinoprostone/metabolism , Glucose/metabolism , Group X Phospholipases A2/genetics , Insulin Secretion , Insulin-Secreting Cells/metabolism , Islets of Langerhans/enzymology , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
Atherosclerosis ; 214(1): 58-64, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20833395

ABSTRACT

OBJECTIVE: Abdominal aortic aneurysm (AAA) is a complex vascular disease characterized by matrix degradation and inflammation and is a major cause of mortality in older men. Specific interventions that prevent AAA progression remain to be identified. In this study, we tested the hypothesis that Group X secretory phospholipase A(2) (GX sPLA(2)), an enzyme implicated in inflammatory processes, mediates AAA. METHODS AND RESULTS: GX sPLA(2) was detected by immunostaining in human aneurysmal tissue and in angiotensin II (Ang II)-induced AAAs in apolipoprotein E-deficient (apoE(-/-)) mice. GX sPLA(2) mRNA was increased significantly (11-fold) in abdominal aortas of apoE(-/-) mice in response to Ang II infusion. To define the role of GX sPLA(2) in experimental AAAs, apoE(-/-) and apoE(-/-) x GX sPLA(2)(-/-) (GX DKO) mice were infused with Ang II for either 10 (n=7) or 28 (n=24-26) days. Deficiency of GX sPLA(2) significantly reduced the incidence and severity of AAAs, as assessed by ultrasound measurements in vivo of aortic lumens and by computer-assisted morphometric analyses ex vivo of external diameter. Results from gene expression profiling indicated that the expression of specific matrix metalloproteinases and inflammatory mediators was blunted in aortas from GX DKO mice compared to apoE(-/-) mice after 10-day Ang II infusion. Ang II induction of cyclooxygenase-2, interleukin-6, matrix metalloproteinase (MMP)-2, MMP-13 and MMP-14 was reduced significantly in GX DKO mice compared to apoE(-/-) mice. CONCLUSION: GX sPLA(2) promotes Ang II-induced pathological responses leading to AAA formation.


Subject(s)
Angiotensin II/metabolism , Aortic Aneurysm, Abdominal/metabolism , Apolipoproteins E/genetics , Group X Phospholipases A2/metabolism , Animals , Aorta, Abdominal/pathology , Apolipoproteins E/metabolism , Blood Pressure , Humans , Inflammation , Interleukins/metabolism , Metalloproteases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic
3.
FASEB J ; 24(11): 4313-24, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20585029

ABSTRACT

Studies in vitro indicate that group X secretory phospholipase A(2) (GX sPLA(2)) potently releases arachidonic acid (AA) and lysophosphatidylcholine from mammalian cell membranes. To define the function of GX sPLA(2) in vivo, our laboratory recently generated C57BL/6 mice with targeted deletion of GX sPLA(2) (GX(-/-) mice). When fed a normal rodent diet, GX(-/-) mice gained significantly more weight and had increased adiposity compared to GX(+/+) mice, which was not attributable to alterations in food consumption or energy expenditure. When treated with adipogenic stimuli ex vivo, stromal vascular cells isolated from adipose tissue of GX(-/-) mice accumulated significantly more (20%) triglyceride compared to cells from GX(+/+) mice. Conversely, overexpression of GX sPLA(2), but not catalytically inactive GX sPLA(2), resulted in a significant 50% reduction in triglyceride accumulation in OP9 adipocytes. The induction of genes encoding adipogenic proteins (PPARγ, SREBP-1c, SCD1, and FAS) was also significantly blunted by 50-80% in OP9 cells overexpressing GX sPLA(2). Activation of the liver X receptor (LXR), a nuclear receptor known to up-regulate adipogenic gene expression, was suppressed in 3T3-L1 and OP9 cells when GX sPLA(2) was overexpressed. Thus, hydrolytic products generated by GX sPLA(2) negatively regulate adipogenesis, possibly by suppressing LXR activation.


Subject(s)
Adipogenesis/physiology , Down-Regulation , Group X Phospholipases A2/metabolism , Adiposity/genetics , Animals , Cell Line , Disease Models, Animal , Gene Expression Regulation , Group X Phospholipases A2/genetics , Humans , Liver X Receptors , Luciferases/metabolism , Male , Mice , Mice, Inbred C57BL , Orphan Nuclear Receptors/metabolism , Triglycerides/metabolism
4.
Arterioscler Thromb Vasc Biol ; 29(4): 532-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19164803

ABSTRACT

OBJECTIVE: In vitro data indicate that human LDL modified by Group V secretory phospholipase A(2) (GV sPLA(2)) is proatherogenic. Consistent with this, gain and loss of function studies demonstrated that GV sPLA(2) promotes atherosclerosis in LDLR(-/-) mice. The current study investigates whether GV sPLA(2) promotes atherosclerotic processes in apoE(-/-) mice. METHODS AND RESULTS: LDL (d=1.019 to 1.063) from apoE(-/-) and LDLR(-/-) mice fed chow or Western diet were hydrolyzed by GV sPLA(2). Phosphatidylcholine on LDL from LDLR(-/-) mice fed either a chow or Western diet was hydrolyzed to a greater extent (61.1+/-0.4% and 45.3+/-4.6%) than the corresponding fractions from apoE(-/-) mice (41.7+/-3.6% and 39.4+/-1.2%). ApoE(-/-) LDL induced macrophage foam cell formation in vitro without modification by GV sPLA(2), whereas hydrolysis of LDLR(-/-) LDL was a prerequisite for foam cell formation. In contrast to findings in LDLR(-/-) mice, GV sPLA(2) deficiency did not significantly reduce atherosclerosis in apoE(-/-) mice, although collagen content was significantly reduced in lesions of apoE(-/-) mice lacking GV sPLA(2). CONCLUSIONS: The ability of GV sPLA(2) to promote atherosclerotic lipid deposition in apoE(-/-) and LDLR(-/-) mice may be related to its ability to increase the atherogenic potential of LDL from these mice as assessed in vitro.


Subject(s)
Aorta/enzymology , Apolipoproteins E/deficiency , Atherosclerosis/enzymology , Group V Phospholipases A2/metabolism , Lipoproteins, LDL/metabolism , Receptors, LDL/deficiency , Animals , Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/etiology , Atherosclerosis/pathology , Cells, Cultured , Collagen/metabolism , Dietary Fats/administration & dosage , Disease Models, Animal , Female , Foam Cells/enzymology , Group V Phospholipases A2/deficiency , Group V Phospholipases A2/genetics , Hydrolysis , Macrophages, Peritoneal/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylcholines/metabolism , Receptors, LDL/genetics , Sphingomyelins/metabolism , Time Factors
5.
J Recept Signal Transduct Res ; 26(1-2): 87-106, 2006.
Article in English | MEDLINE | ID: mdl-16595340

ABSTRACT

Few data exist on the modulation of cytokine receptor signaling by the actin or tubulin cytoskeleton. Therefore, we studied interleukin-2 receptor (IL-2R) signaling in phytohemagglutinine (PHA)-pretreated human T cells in the context of alterations in the cytoskeletal system induced by cytochalasin D (CyD), jasplaklinolide (Jas), taxol (Tax), or colchicine (Col). We found that changes in cytoskeletal tubulin polymerization altered the strength of several IL-2-triggered signals. Moreover, Tax-induced tubulin hyperpolymerization augmented the surface expression of the IL-2R ss -chain and enhanced the association of the IL-2R beta -chain with cytoskeletal tubulin. The IL-2R beta-chain, in turn, was constitutively associated with tubulin and, more weakly, actin. To exclude the possibility that these associations are artifacts caused by PHA, we confirmed them in T cells from TCR-transgenic DO 11.10 mice stimulated with their nominal antigen. We conclude that altered polymerization of cytoskeletal components, especially tubulin, is accompanied by modulation of IL-2 signaling at the receptor level.


Subject(s)
Receptors, Interleukin-2/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Tubulin/metabolism , Actins/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Colchicine/pharmacology , Cytochalasin D/pharmacology , Cytoskeleton/metabolism , Depsipeptides/pharmacology , Gout Suppressants/pharmacology , Humans , Interleukin-2/pharmacology , Mice , Mice, Transgenic , Mitogens/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , Paclitaxel/pharmacology , Phytohemagglutinins/pharmacology
6.
J Investig Med ; 53(6): 322-8, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16207471

ABSTRACT

Although the immunomodulatory properties of statins are in part independent of their lipid-lowering effects, cholesterol is a major component of lipid rafts. We therefore studied the effects of atorvastatin (AS) on the raft enrichment of the interleukin-2 receptor (IL-2R) beta chain previously described by us and on early IL-2R signaling events in activated human T cells. We found that concomitant AS exposure during a 3-day stimulation with phytohemagglutinin (PHA) attenuates activation-associated events, such as the enhanced surface expression of the raft marker GM-1 and the induced expression of the activation marker CD25 (the IL-2R alpha chain). In contrast, brief AS treatment after PHA stimulation increased GM-1 surface expression and virtually abolished the selective raft enrichment of the IL-2R beta chain. Although this AS-associated increase in GM-1 expression resembled that seen in the presence of the raft-disrupting cholesterol chelator methyl-beta-cyclodextrin (MBCD), the two agents had contrasting effects on the tyrosine phosphorylation of the IL-2R beta chain by exogenous IL-2: MBCD essentially abolished this event, whereas AS tended to enhance it and shifted its occurrence out of rafts. We conclude that AS affects IL-2R signaling by altering the raft enrichment of the IL-2R beta chain and propose that this effect is one mechanism underlying the immunomodulatory properties of statins.


Subject(s)
Heptanoic Acids/pharmacology , Immunologic Factors/pharmacology , Interleukin-2/metabolism , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Pyrroles/pharmacology , Receptors, Interleukin/drug effects , Receptors, Interleukin/metabolism , Anticholesteremic Agents/pharmacology , Atorvastatin , Humans , In Vitro Techniques , Interleukin-2 Receptor beta Subunit , Signal Transduction/drug effects
7.
Transpl Immunol ; 14(1): 43-7, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15814281

ABSTRACT

Stimulated human T cells from healthy volunteers demonstrate attenuated early interleukin (IL)-2 receptor (R) signaling in the presence of daclizumab (Dac). Aiming to confirm that this ex-vivo effect of Dac is also observed in-vivo, we studied T cells from 3 kidney transplant recipients before and 2-3 weeks and 4-6 months after transplantation. We found by flow cytometry that T cells obtained pre-transplant and stimulated ex-vivo with phytohemeagglutinine upregulated the IL-2R alpha-(CD25) and beta-(CD122) chains as expected. Moreover, exogenous IL-2 induced characteristic tyrosine phosphorylation events detectable by immunoblotting in these cells. However, T cells studied post-transplant neither exhibited CD25 or -122 upregulation nor IL-2-induced tyrosine phosphorylation events, indicating broad, persistent suppression of the IL-2R signaling machinery which thus appears largely inaccessible for Dac in actual transplant recipients. We therefore conclude that the clinical efficacy of this agent may depend on additional mechanisms in-vivo other than those identified ex-vivo.


Subject(s)
Antibodies, Monoclonal/pharmacology , Immunoglobulin G/pharmacology , Kidney Transplantation , Receptors, Interleukin-2/metabolism , T-Lymphocytes/drug effects , Adolescent , Age Factors , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Daclizumab , Down-Regulation , Humans , Immunoglobulin G/therapeutic use , Signal Transduction , T-Lymphocytes/immunology
8.
Nurs Econ ; 22(3): 124-34, 107, 2004.
Article in English | MEDLINE | ID: mdl-15211915

ABSTRACT

Financial performance measures are essential to improve the fiscal management of academic nurse-managed centers (ANMCs). Measures are compared among six ANMCs in a consortium and against an external, self-sustainable, profitable ANMC and national data for family practice physicians. Performance measures help identify a center's strengths and weaknesses facilitating the development of strategies aimed at a variety of targets (business practices related to revenue and costs) to improve financial viability. Using a variety of financial performance measures to inform decision making will aid ANMCs in keeping their doors open for business.


Subject(s)
Academic Medical Centers/organization & administration , Ambulatory Care Facilities/organization & administration , Efficiency, Organizational/economics , Financial Audit , Nurse Administrators , Nurse Clinicians/organization & administration , Academic Medical Centers/economics , Academic Medical Centers/statistics & numerical data , Ambulatory Care Facilities/economics , Ambulatory Care Facilities/statistics & numerical data , Capitation Fee , Cost Allocation , Current Procedural Terminology , Data Collection , Fee-for-Service Plans , Humans , Michigan , Pilot Projects , Professional Autonomy , Relative Value Scales , Texas
9.
Cytokine Growth Factor Rev ; 15(2-3): 103-10, 2004.
Article in English | MEDLINE | ID: mdl-15110794

ABSTRACT

Lipid rafts are established as critical structures for a variety of cellular processes, including immune cell activation. Beyond their importance for initial immune cell activation at the immunological synapse, lipid rafts are now also being recognized as important sites for cytokine and growth factor signal transduction, both in immune cells as part of secondary regulatory processes, and in non-immune cells. This review summarizes current knowledge regarding the roles of rafts in cytokine signaling and emphasizes the need for measures to better standardize the study of rafts.


Subject(s)
Cytokines/physiology , Membrane Microdomains/physiology , Signal Transduction/physiology , Humans , Membrane Microdomains/ultrastructure
10.
Hum Immunol ; 63(10): 813-20, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12368033

ABSTRACT

Glycolipid-enriched membrane microdomains ("rafts") are critical sites for signal transduction and other processes such as intracellular transport. While the participation of T-cell rafts in the formation of the immunological synapse is well established, the role of rafts on antigen-presenting cells (APCs) as well as the relationship between these domains and major histocompatibility complex (MHC) molecules is less clearly defined. We therefore investigated whether MHC class I or II molecules are found in rafts of the human macrophage-monocytic cell line U937. We detected the preferential localization of MHC class II, but not class I, molecules in rafts. Furthermore, raft disruption resulted in a decrease in constitutive protein tyrosine phosphorylation events in U937 cells. Our findings are reviewed in the context of results from other groups who also found important associations of MHC class II molecules with APC rafts. Additional, and at times contradictory, findings by others regarding the relationship between rafts and MHC molecules are also discussed. It is concluded that class II MHC molecules can localize in rafts of APCs and that this localization may be relevant for APC function and thus immune regulation.


Subject(s)
Antigen-Presenting Cells/immunology , Histocompatibility Antigens Class II/analysis , Histocompatibility Antigens Class I/analysis , Membrane Microdomains/physiology , Antigens, Differentiation, B-Lymphocyte/analysis , G(M1) Ganglioside/analysis , Humans , Phosphorylation , Tyrosine/metabolism , U937 Cells , beta 2-Microglobulin/analysis
11.
Biochem Biophys Res Commun ; 295(2): 540-6, 2002 Jul 12.
Article in English | MEDLINE | ID: mdl-12150984

ABSTRACT

Calpain, a calcium-dependent cysteine protease, is known to associate with the T-cell plasma membrane and subsequently cleave a number of cytoskeletal-associated proteins. In this study, we report the novel observation that calpain II, but not calpain I, associates with membrane lipid rafts on human peripheral blood T-cells and Jurkat cells. Raft-associated calpain activity is enhanced with exogenous calcium and inhibited with calpeptin, a specific inhibitor of calpain activity. In addition, we demonstrate that calpain cleaves the cytoskeletal-associated protein, talin, during the first 30-min after cell stimulation. We propose that lipid raft associated-calpain II could function in early TCR signaling to facilitate immune synapse formation through cytoskeletal remodeling mechanisms. Hence, we demonstrate that the positioning of calpain II within T-cell lipid rafts strategically places it in close proximity to known calpain substrates that are cleaved during Ag-specific T-cell signaling and immune synapse formation.


Subject(s)
Calpain/metabolism , Membrane Lipids/metabolism , T-Lymphocytes/enzymology , Detergents , Humans , Jurkat Cells , T-Lymphocytes/metabolism
12.
J Leukoc Biol ; 72(1): 199-206, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12101281

ABSTRACT

We studied whether cytokine receptors (Rs) on T cells associate with lipid microdomains ("rafts"). Low-dose phytohemagglutinin (PHA)-stimulated human T cells were separated into cytoplasmic, membrane, and raft fractions by buoyant density centrifugation. Examination of these fractions for the presence of interleukin (IL)-2- and -15R chains and associated signaling molecules by Western blotting revealed marked, selective enrichment of the IL-2/15R beta-chain in rafts before IL-2 stimulation. After IL-2 stimulation, a substantial amount of the beta-chain was found in the membrane fraction. This partial translocation was also observed for the beta-chain-associated molecules JAK-1, p56(lck), and grb-2. Finally, raft disruption with methyl-beta-cyclodextrin (MBCD) attenuated IL-2-induced tyrosine phosphorylation events and selectively decreased the surface expression of the IL-2/15R beta-chain detected by flow cytometry. These results show that the IL-2/15R beta-chain is enriched in rafts obtained from low-dose, PHA-stimulated T cells, that IL-2 binding alters this enrichment, and that this enrichment may be functionally relevant as a possible mechanism to ensure cytokine selectivity and specificity.


Subject(s)
Membrane Microdomains/chemistry , Receptors, Interleukin-2/analysis , Receptors, Interleukin/analysis , T-Lymphocytes/immunology , beta-Cyclodextrins , Cells, Cultured , Cyclodextrins/pharmacology , G(M1) Ganglioside/analysis , Humans , Interleukin-2/pharmacology , Interleukin-2 Receptor beta Subunit , Lymphocyte Activation , Membrane Microdomains/drug effects , Membrane Microdomains/physiology , Protein Subunits , Receptors, Interleukin-15 , Signal Transduction , T-Lymphocytes/chemistry , T-Lymphocytes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...