Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
FASEB J ; 32(8): 4121-4131, 2018 08.
Article in English | MEDLINE | ID: mdl-29509511

ABSTRACT

Lipid metabolism is closely involved with signal transduction and energy homeostasis. Excess calorie intake causes abnormal lipid metabolism, promoting obesity and insulin resistance. Diacylglycerol (DG) represents not only a lipidic second messenger but also an intermediate metabolite for triglyceride metabolism in the endoplasmic reticulum (ER). However, it remains undetermined how the roles of DG in signaling and energy homeostasis is regulated within the cell. Of DG kinases (DGKs), which are enzymes that phosphorylate DG, DGKε resides in the ER. This study examined how DGKε is implicated in signal transduction and lipid homeostasis. DGKε-deficient mice were fed a high-fat diet (HFD) for 40 d. We observed that DGKε deficiency promotes fat accumulation in adipocytes and subsequently promotes insulin resistance in mice fed an HFD. This abnormal fat metabolism is mediated by down-regulation of lipolytic activities, such as adipose triglyceride lipase and hormone-sensitive lipase. In addition, activation of DG-sensitive PKC leads to insulin resistance in adipose tissue, which may be caused by delayed metabolism of DG. Our data suggest that DGKε links the second messenger signaling system to energy homeostasis in adipocytes and that its deficiency results in abnormal lipid metabolism such as obesity and insulin resistance.-Nakano, T., Seino, K., Wakabayashi, I., Stafforini, D. M., Topham, M. K., Goto, K. Deletion of diacylglycerol kinase ε confers susceptibility to obesity via reduced lipolytic activity in murine adipocytes.


Subject(s)
Adipocytes/metabolism , Diacylglycerol Kinase/metabolism , Obesity/metabolism , Animals , Diet, High-Fat/adverse effects , Down-Regulation/physiology , Homeostasis/physiology , Insulin Resistance/physiology , Lipase/metabolism , Lipid Metabolism/physiology , Mice , Signal Transduction/physiology
2.
Semin Cancer Biol ; 35 Suppl: S151-S184, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25951989

ABSTRACT

Cancers harbor significant genetic heterogeneity and patterns of relapse following many therapies are due to evolved resistance to treatment. While efforts have been made to combine targeted therapies, significant levels of toxicity have stymied efforts to effectively treat cancer with multi-drug combinations using currently approved therapeutics. We discuss the relationship between tumor-promoting inflammation and cancer as part of a larger effort to develop a broad-spectrum therapeutic approach aimed at a wide range of targets to address this heterogeneity. Specifically, macrophage migration inhibitory factor, cyclooxygenase-2, transcription factor nuclear factor-κB, tumor necrosis factor alpha, inducible nitric oxide synthase, protein kinase B, and CXC chemokines are reviewed as important antiinflammatory targets while curcumin, resveratrol, epigallocatechin gallate, genistein, lycopene, and anthocyanins are reviewed as low-cost, low toxicity means by which these targets might all be reached simultaneously. Future translational work will need to assess the resulting synergies of rationally designed antiinflammatory mixtures (employing low-toxicity constituents), and then combine this with similar approaches targeting the most important pathways across the range of cancer hallmark phenotypes.


Subject(s)
Antineoplastic Agents/therapeutic use , Inflammation/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Cell Transformation, Neoplastic/drug effects , Genetic Heterogeneity/drug effects , Humans , Inflammation/genetics , Inflammation/pathology , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/pathology , Signal Transduction/drug effects
3.
J Immunol ; 191(4): 1907-15, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23851686

ABSTRACT

IFN-ε is a unique type I IFN whose constitutive expression in lung, brain, small intestine, and reproductive tissues is only partially understood. Our previous observation that posttranscriptional events participate in the regulation of IFN-ε mRNA expression led us to investigate whether the 5' and/or 3' untranslated regions (UTR) have regulatory functions. Surprisingly, we found that full-length IFN-ε 5'UTR markedly suppressed mRNA expression under basal conditions. Analysis of the secondary structure of this region predicted formation of two stable stem-loop structures, loops 1 and 2. Studies using luciferase constructs harboring various stretches of IFN-ε 5'UTR and mutant constructs in which the conformation of loop structures was disrupted showed that loop 1 is essential for regulation of mRNA expression. Incubation of HeLa cell extracts with agarose-bound RNAs harboring IFN-ε loop structures identified importin 9 (IPO9), a molecular transporter and chaperone, as a candidate that associates with these regions of the 5'UTR. IPO9 overexpression decreased, and IPO9 silencing increased basal IFN-ε expression. Our studies uncover a previously undescribed function for IPO9 as a specific, and negative, posttranscriptional regulator of IFN-ε expression, and they identify key roles for IFN-ε stem-loop structure 1 in this process. IPO9-mediated effects on 5'UTRs appear to extend to additional mRNAs, including hypoxia-inducible factor-1α, that can form specific loop structures.


Subject(s)
5' Untranslated Regions/genetics , Gene Expression Regulation/physiology , Interferons/genetics , Inverted Repeat Sequences/genetics , Karyopherins/physiology , RNA Interference , Animals , Base Sequence , Consensus Sequence , Down-Regulation , Female , Genes, Reporter , HeLa Cells , Humans , Interferons/biosynthesis , Karyopherins/genetics , Mammals/genetics , Molecular Sequence Data , Nucleic Acid Conformation , RNA Stability , RNA, Messenger/biosynthesis , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Homology, Nucleic Acid , Uterine Cervical Neoplasms/pathology
4.
J Lipid Res ; 55(8): 1657-67, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24879802

ABSTRACT

ACSL4 is a member of the long-chain acyl-CoA synthetase (ACSL) family with a marked preference for arachidonic acid (AA) as its substrate. Although an association between elevated levels of ACSL4 and hepatosteatosis has been reported, the function of ACSL4 in hepatic FA metabolism and the regulation of its functional expression in the liver remain poorly defined. Here we provide evidence that AA selectively downregulates ACSL4 protein expression in hepatic cells. AA treatment decreased the half-life of ACSL4 protein in HepG2 cells by approximately 4-fold (from 17.3 ± 1.8 h to 4.2 ± 0.4 h) without causing apoptosis. The inhibitory action of AA on ACSL4 protein stability could not be prevented by rosiglitazone or inhibitors that interfere with the cellular pathways involved in AA metabolism to biologically active compounds. In contrast, treatment of cells with inhibitors specific for the proteasomal degradation pathway largely prevented the AA-induced ACSL4 degradation. We further show that ACSL4 is intrinsically ubiquitinated and that AA treatment can enhance its ubiquitination. Collectively, our studies have identified a novel substrate-induced posttranslational regulatory mechanism by which AA downregulates ACSL4 protein expression in hepatic cells.


Subject(s)
Arachidonic Acid/pharmacology , Coenzyme A Ligases/biosynthesis , Down-Regulation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Hepatocytes/enzymology , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Ubiquitination/drug effects , Animals , Coenzyme A Ligases/genetics , Hep G2 Cells , Humans , Male , Mice , Proteasome Endopeptidase Complex/genetics
5.
J Biol Chem ; 288(3): 1439-47, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23184957

ABSTRACT

Discovering proteins that modulate Akt signaling has become a critical task, given the oncogenic role of Akt in a wide variety of cancers. We have discovered a novel diacylglycerol signaling pathway that promotes dephosphorylation of Akt. This pathway is regulated by diacylglycerol kinase δ (DGKδ). In DGKδ-deficient cells, we found reduced Akt phosphorylation downstream of three receptor tyrosine kinases. Phosphorylation upstream of Akt was not affected. Our data indicate that PKCα, which is excessively active in DGKδ-deficient cells, promotes dephosphorylation of Akt through pleckstrin homology domain leucine-rich repeats protein phosphatase (PHLPP) 2. Depletion of either PKCα or PHLPP2 rescued Akt phosphorylation in DGKδ-deficient cells. In contrast, depletion of PHLPP1, another Akt phosphatase, failed to rescue Akt phosphorylation. Other PHLPP substrates were not affected by DGKδ deficiency, suggesting mechanisms allowing specific modulation of Akt dephosphorylation. We found that ß-arrestin 1 acted as a scaffold for PHLPP2 and Akt1, providing a mechanism for specificity. Because of its ability to reduce Akt phosphorylation, we tested whether depletion of DGKδ could attenuate tumorigenic properties of cultured cells and found that DGKδ deficiency reduced cell proliferation and migration and enhanced apoptosis. We have, thus, discovered a novel pathway in which diacylglycerol signaling negatively regulates Akt activity. Our collective data indicate that DGKδ is a pertinent cancer target, and our studies could lay the groundwork for development of novel cancer therapeutics.


Subject(s)
Diacylglycerol Kinase/metabolism , Diglycerides/metabolism , Phosphoprotein Phosphatases/deficiency , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Arrestins/genetics , Arrestins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic , Diacylglycerol Kinase/genetics , Gene Expression Regulation , Gene Knockdown Techniques , Genetic Vectors , Humans , Lentivirus/genetics , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Phosphoprotein Phosphatases/genetics , Phosphorylation , Protein Kinase C-alpha/deficiency , Protein Kinase C-alpha/genetics , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/genetics , Substrate Specificity , beta-Arrestin 1 , beta-Arrestins
6.
J Lipid Res ; 53(9): 1767-82, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22665167

ABSTRACT

Lipoprotein-associated phospholipase A(2) (Lp-PLA(2)), also known as platelet-activating factor acetylhydrolase (PAF-AH), is a unique member of the phospholipase A(2) superfamily. This enzyme is characterized by its ability to specifically hydrolyze PAF as well as glycerophospholipids containing short, truncated, and/or oxidized fatty acyl groups at the sn-2 position of the glycerol backbone. In humans, Lp-PLA(2) circulates in active form as a complex with low- and high-density lipoproteins. Clinical studies have reported that plasma Lp-PLA(2) activity and mass are strongly associated with atherogenic lipids and vascular risk. These observations led to the hypothesis that Lp-PLA(2) activity and/or mass levels could be used as biomarkers of cardiovascular disease and that inhibition of the activity could offer an attractive therapeutic strategy. Darapladib, a compound that inhibits Lp-PLA(2) activity, is anti-atherogenic in mice and other animals, and it decreases atherosclerotic plaque expansion in humans. However, disagreement continues to exist regarding the validity of Lp-PLA(2) as an independent marker of atherosclerosis and a scientifically justified target for intervention. Circulating Lp-PLA(2) mass and activity are associated with vascular risk, but the strength of the association is reduced after adjustment for basal concentrations of the lipoprotein carriers with which the enzyme associates. Genetic studies in humans harboring an inactivating mutation at this locus indicate that loss of Lp-PLA(2) function is a risk factor for inflammatory and vascular conditions in Japanese cohorts. Consistently, overexpression of Lp-PLA(2) has anti-inflammatory and anti-atherogenic properties in animal models. This thematic review critically discusses results from laboratory and animal studies, analyzes genetic evidence, reviews clinical work demonstrating associations between Lp-PLA(2) and vascular disease, and summarizes results from animal and human clinical trials in which administration of darapladib was tested as a strategy for the management of atherosclerosis.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Atherosclerosis/enzymology , Inflammation/enzymology , Oxidative Stress , 1-Alkyl-2-acetylglycerophosphocholine Esterase/antagonists & inhibitors , 1-Alkyl-2-acetylglycerophosphocholine Esterase/chemistry , 1-Alkyl-2-acetylglycerophosphocholine Esterase/deficiency , 1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Animals , Atherosclerosis/drug therapy , Atherosclerosis/epidemiology , Atherosclerosis/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Inflammation/drug therapy , Inflammation/epidemiology , Inflammation/metabolism , Oxidative Stress/drug effects
7.
J Biol Chem ; 285(10): 6952-9, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20064931

ABSTRACT

Many human epithelial cancers are characterized by abnormal activation of the epidermal growth factor receptor (EGFR), which is often caused by its excessive expression in tumor cells. The abundance of EGFR is modulated, in part, by its ubiquitination, which targets it for degradation. The components responsible for adding ubiquitin to EGFR are well characterized, but this is a reversible process, and the mechanisms that modulate the removal of ubiquitin from the EGFR are not well known. We found that de-ubiquitination of EGFR was regulated by diacylglycerol kinase delta (DGKdelta), a lipid kinase that terminates diacylglycerol signaling. In DGKdelta-deficient cells, ubiquitination of EGFR was enhanced, which attenuated the steady-state levels of EGFR and promoted its ligand-induced degradation. These effects were not caused by changes in the ubiquitinating apparatus, but instead were due to reduced expression of the de-ubiquitinase, ubiquitin-specific protease 8 (USP8). Depletion of protein kinase Calpha (PKCalpha), a target of diacylglycerol, rescued the levels of USP8 and normalized EGFR degradation in DGKdelta-deficient cells. Moreover, the effects of PKCalpha were caused by its inhibition of Akt, which stabilizes USP8. Our data indicate a novel mechanism where DGKdelta and PKCalpha modulate the levels of ubiquitinated EGFR through Akt and USP8.


Subject(s)
Diacylglycerol Kinase/metabolism , Endopeptidases/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , ErbB Receptors/metabolism , Protein Kinase C-alpha/metabolism , Ubiquitin Thiolesterase/metabolism , Animals , Cell Line, Tumor , Diacylglycerol Kinase/genetics , Endocytosis/physiology , Endopeptidases/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Enzyme Activation , ErbB Receptors/genetics , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Myristoylated Alanine-Rich C Kinase Substrate , Protein Kinase C-alpha/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-cbl/metabolism , RNA Interference , Ubiquitin Thiolesterase/genetics , Ubiquitinated Proteins/genetics , Ubiquitinated Proteins/metabolism , Ubiquitination
8.
Am J Physiol Heart Circ Physiol ; 301(6): H2395-401, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21949111

ABSTRACT

Sodium-hydrogen exchanger (NHE), the principal sarcolemmal acid extruder in ventricular myocytes, is stimulated by a variety of autocrine/paracrine factors and contributes to myocardial injury and arrhythmias during ischemia-reperfusion. Platelet-activating factor (PAF; 1-o-alkyl-2-acetyl-sn-glycero-3-phosphocholine) is a potent proinflammatory phospholipid that is released in the heart in response to oxidative stress and promotes myocardial ischemia-reperfusion injury. PAF stimulates NHE in neutrophils and platelets, but its effect on cardiac NHE (NHE1) is unresolved. We utilized quiescent guinea pig ventricular myocytes bathed in bicarbonate-free solutions and epifluorescence to measure intracellular pH (pH(i)). Methylcarbamyl-PAF (C-PAF; 200 nM), a metabolically stable analog of PAF, significantly increased steady-state pH(i). The alkalosis was completely blocked by the NHE inhibitor, cariporide, and by sodium-free bathing solutions, indicating it was mediated by NHE activation. C-PAF also significantly increased the rate of acid extrusion induced by intracellular acidosis. The ability of C-PAF to increase steady-state pH(i) was completely blocked by the PAF receptor inhibitor WEB 2086 (10 µM), indicating the PAF receptor is required. A MEK inhibitor (PD98059; 25 µM) also completely blocked the rise in pH(i) induced by C-PAF, suggesting participation of the MAP kinase signaling cascade downstream of the PAF receptor. Inhibition of PKC with GF109203X (1 µM) and chelerythrine (2 µM) did not significantly affect the alkalosis induced by C-PAF. In summary, these results provide evidence that PAF stimulates cardiac NHE1, the effect occurs via the PAF receptor, and signal relay requires participation of the MAP kinase cascade.


Subject(s)
Heart Ventricles/metabolism , Myocytes, Cardiac/metabolism , Platelet Activating Factor/metabolism , Sodium-Hydrogen Exchangers/metabolism , Sodium/metabolism , Acidosis/metabolism , Alkalosis/metabolism , Animals , Azepines/pharmacology , Benzophenanthridines/pharmacology , Dose-Response Relationship, Drug , Flavonoids/pharmacology , Guanidines/pharmacology , Guinea Pigs , Heart Ventricles/drug effects , Hydrogen-Ion Concentration , Indoles/pharmacology , Ion Transport , Maleimides/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Myocytes, Cardiac/drug effects , Phospholipid Ethers/pharmacology , Platelet Membrane Glycoproteins/drug effects , Platelet Membrane Glycoproteins/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Spectrometry, Fluorescence , Sulfones/pharmacology , Time Factors , Triazoles/pharmacology
9.
Blood ; 113(26): 6699-706, 2009 Jun 25.
Article in English | MEDLINE | ID: mdl-19395675

ABSTRACT

Platelet activating factor (PAF) and PAF-like lipids induce inflammatory responses in target cells. These lipid mediators are inactivated by PAF-acetylhydrolase (PAF-AH). The PAF signaling system affects the growth of hematopoietic CD34(+) cells, but roles for PAF-AH in this process are unknown. Here, we investigated PAF-AH function during megakaryopoiesis and found that human CD34(+) cells accumulate this enzymatic activity as they differentiate toward megakaryocytes, consistent with the expression of mRNA and protein for the plasma PAF-AH isoform. Inhibition of endogenous PAF-AH activity in differentiated megakaryocytes increased formation of lipid mediators that signaled the PAF receptor (PAFR) in fully differentiated human cells such as neutrophils, as well as megakaryocytes themselves. PAF-AH also controlled megakaryocyte alpha(IIb)beta(3)-dependent adhesion, cell spreading, and mobility that relied on signaling through the PAFR. Together these data suggest that megakaryocytes generate PAF-AH to modulate the accumulation of intracellular phospholipid mediators that may detrimentally affect megakaryocyte development and function.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/physiology , Megakaryocytes/metabolism , Phospholipids/metabolism , Thrombopoiesis/physiology , 1-Alkyl-2-acetylglycerophosphocholine Esterase/biosynthesis , 1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Calcium Signaling , Cell Adhesion , Cells, Cultured/cytology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Enzyme Induction , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/enzymology , Humans , Interleukin-3/pharmacology , Megakaryocytes/cytology , Megakaryocytes/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoproteins/physiology , RNA, Messenger/biosynthesis , Receptors, G-Protein-Coupled/physiology , Stem Cell Factor/pharmacology , Thrombopoiesis/drug effects , Thrombopoietin/pharmacology
10.
Crit Rev Immunol ; 30(6): 489-513, 2010.
Article in English | MEDLINE | ID: mdl-21175414

ABSTRACT

Antiviral innate immunity is triggered by sensing viral nucleic acids. RIG-I (retinoic acid-inducible gene-I) is an intracellular molecule that responds to viral nucleic acids and activates downstream signaling, resulting in the induction of members of the type I interferon (IFN) family, which are regarded among the most important effectors of the innate immune system. Although RIG-I is expressed ubiquitously in the cytoplasm, its levels are subject to transcriptional and post-transcriptional regulation. RIG-I belongs to the IFN-stimulated gene (ISG) family, but certain cells regulate its expression through IFN-independent mechanisms. Several lines of evidence indicate that deregulated RIG-I signaling is associated with autoimmune disorders. Further studies suggest that RIG-I has functions in addition to those directly related to its role in RNA sensing and host defense. We have much to learn and discover regarding this interesting cytoplasmic sensor so that we can capitalize on its properties for the treatment of viral infections, immune disorders, cancer, and perhaps other conditions.


Subject(s)
DEAD-box RNA Helicases/immunology , Animals , DEAD-box RNA Helicases/metabolism , Humans , Interferons/immunology , Ligands , Signal Transduction , Ubiquitin/immunology , Ubiquitin/metabolism
11.
J Immunol ; 182(5): 2717-25, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19234166

ABSTRACT

Retinoic acid-inducible gene I (RIG-I) is an intracellular pattern recognition receptor that plays important roles during innate immune responses to viral dsRNAs. The mechanisms and signaling molecules that participate in the downstream events that follow activation of RIG-I are incompletely characterized. In addition, the factors that define intracellular availability of RIG-I and determine its steady-state levels are only partially understood but are likely to play a major role during innate immune responses. It was recently reported that the antiviral activity of RIG-I is negatively regulated by specific E3 ubiquitin ligases, suggesting participation of the proteasome in the regulation of RIG-I levels. In this study, we used immunoprecipitation combined with mass spectrometry to identify RIG-I-interacting proteins and found that RIG-I forms part of a protein complex that includes heat shock protein 90-alpha (HSP90-alpha), a molecular chaperone. Biochemical studies using purified systems demonstrated that the association between RIG-I and HSP90-alpha is direct but does not involve participation of the CARD domain. Inhibition of HSP90 activity leads to the dissociation of the RIG-I-HSP90 complex, followed by ubiquitination and proteasomal degradation of RIG-I. In contrast, the levels of RIG-I mRNA are unaffected. Our studies also show that the ability of RIG-I to respond to stimulation with polyinosinic:polycytidylic acid is abolished when its interaction with HSP90 is inhibited. These novel findings point to HSP90-alpha as a chaperone that shields RIG-I from proteasomal degradation and modulates its activity. These studies identify a new mechanism whose dysregulation may seriously compromise innate antiviral responses in mammals.


Subject(s)
DEAD-box RNA Helicases/antagonists & inhibitors , DEAD-box RNA Helicases/metabolism , HSP90 Heat-Shock Proteins/physiology , Adenocarcinoma/enzymology , Adenocarcinoma/metabolism , Amino Acid Sequence , Cell Line , Colonic Neoplasms/enzymology , Colonic Neoplasms/metabolism , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/physiology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , HT29 Cells , HeLa Cells , Humans , Molecular Sequence Data , Proteasome Endopeptidase Complex/metabolism , Protein Interaction Mapping , Protein Structure, Tertiary/genetics , Receptors, Immunologic , Transcriptional Activation/immunology , Ubiquitination/immunology
12.
Biochem J ; 428(2): 269-79, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20331434

ABSTRACT

The plasma form of PAF-AH [PAF (platelet-activating factor) acetylhydrolase; also known as LpPLA(2) (lipopoprotein-associated phospholipase A(2)), PLA(2)G7] catalyses the release of sn-2 fatty acyl residues from PAF, oxidatively fragmented phospholipids, and esterified isoprostanes. The plasma levels of this enzyme vary widely among mammalian species, including mice and humans, but the mechanisms that account for these differences are largely unknown. We investigated the basis for these variations using molecular and biochemical approaches. We identified an N-terminal domain that played key roles in the determination of steady-state expression levels. The mouse N-terminal domain robustly enhanced protein expression levels, possibly owing to its ability to adopt a globular conformation that is absent in the human protein. We investigated the mechanism(s) whereby the N-terminal stretch modulated PAF-AH levels and found that differential expression was not due to variations in the efficiency of transcription, translation, or mRNA stability. Studies designed to evaluate the ability of precursor forms of PAF-AH to mature to fully active proteins indicated that the N-terminal end of human and mouse PAF-AH played important and opposite roles in this process. These domains also modulated the levels of expression of an unrelated polypeptide by affecting the stability of precursor forms of the protein. These studies provide insights that contribute to our understanding of the molecular features and mechanisms that contribute to differential expression of plasma PAF-AH in mammals.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/blood , 1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase/chemistry , 1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , HCT116 Cells , HT29 Cells , Humans , Mice , Molecular Sequence Data , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , RNA Stability , Reverse Transcriptase Polymerase Chain Reaction
13.
Pediatr Res ; 68(3): 225-30, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20531249

ABSTRACT

Human preterm infants with necrotizing enterocolitis (NEC) have increased circulating and luminal levels of platelet-activating factor (PAF) and decreased serum PAF-acetylhydrolase (PAF-AH), the enzyme that inactivates PAF. Formula supplemented with recombinant PAF-AH decreases NEC in a neonatal rat model. We hypothesized that endogenous PAF-AH contributes to neonatal intestinal homeostasis and therefore developed PAF-AH mice using standard approaches to study the role of this enzyme in the neonatal NEC model. After exposure to a well-established NEC model, intestinal tissues were evaluated for histology, proinflammatory cytokine mRNA synthesis, and death using standard techniques. We found that mortality rates were significantly lower in PAF-AH pups compared with wild-type controls before 24 h of life but surviving PAF-AH animals were more susceptible to NEC development compared with wild-type controls. Increased NEC incidence was associated with prominent inflammation characterized by elevated intestinal mRNA expression of sPLA2, inducible NOS, and CXCL1. In conclusion, the data support a protective role for endogenous PAF-AH in the development of NEC, and because preterm neonates have endogenous PAF-AH deficiency, this may place them at increased risk for disease.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/deficiency , Enterocolitis, Necrotizing/enzymology , RNA, Messenger/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase/blood , 1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Analysis of Variance , Animals , Apoptosis/physiology , Chemokine CXCL1/metabolism , Cytokines/metabolism , Enterocolitis, Necrotizing/mortality , Enterocolitis, Necrotizing/pathology , Mice , Mice, Knockout , Nitric Oxide Synthase Type II/metabolism , Phospholipases A2, Secretory/metabolism , Platelet Activating Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction
14.
Int J Mol Sci ; 11(12): 5339-47, 2010.
Article in English | MEDLINE | ID: mdl-21614211

ABSTRACT

We previously showed that plasminogen (Plg) isolated from the plasma of normal human subjects contains 1-2 moles of oxidized phosphatidylcholine (oxPtdPC) adducts/mole of protein. Moreover, we suggested that these species are generated at the hepatic site and speculated that they may play a role in the reported cardiovascular pathogenicity of Plg. We aimed to determine whether mouse Plg also harbors linked oxPtdPCs and whether these molecules are metabolized by lipoprotein-associated phospholipase A(2)/PAF acetylhydrolase (Lp-PLA(2)/PAF-AH), an enzyme specific for hydrolysis of oxPtdPCs. We determined the total concentration of Plg in plasma samples from control (WT) and Lp-PLA(2)-deficient (KO) mice, we isolated Plg, and assessed its content of oxPtdPCs by immunoblot analyses. We also evaluated whether human recombinant Lp-PLA(2) metabolized Plg-linked oxPtdPCs in vivo and in vitro. WT and KO mice expressed comparable levels (14.4-15.8 mg/dL) of plasma Plg, as determined by ELISA. We observed no differences in the content of oxPtdPC in Plg isolated from the two mouse strains and in parallel no changes in oxPtdPC content in mouse Plg following incubation with pure recombinant Lp-PLA(2). Plg from mouse plasma contains oxPtdPC adducts that are not affected by the action of Lp-PLA(2), suggesting that linkage to Plg protects oxPtdPCs from metabolism during their transport in the plasma. This modification may have important physio-pathological implications related to the function of Plg, oxPtdPCs, or both.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/chemistry , Phosphatidylcholines/chemistry , Plasminogen/chemistry , 1-Alkyl-2-acetylglycerophosphocholine Esterase/blood , Animals , Humans , Mice , Mice, Knockout , Oxidation-Reduction , Phosphatidylcholines/blood , Phosphatidylcholines/isolation & purification , Plasminogen/isolation & purification , Plasminogen/metabolism
16.
Cardiovasc Drugs Ther ; 23(1): 73-83, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18949548

ABSTRACT

INTRODUCTION: This article is focused on platelet-activating factor acetylhydrolase (PAF-AH), a lipoprotein bound, calcium-independent phospholipase A(2) activity also referred to as lipoprotein-associated phospholipase A(2) or PLA(2)G7. PAF-AH catalyzes the removal of the acyl group at the sn-2 position of PAF and truncated phospholipids generated in settings of inflammation and oxidant stress. DISCUSSION: Here, I discuss current knowledge related to the structural features of this enzyme, including the molecular basis for association with lipoproteins and susceptibility to oxidative inactivation. The circulating form of PAF-AH is constitutively active and its expression is upregulated by mediators of inflammation at the transcriptional level. This mechanism is likely responsible for the observed up-regulation of PAF-AH during atherosclerosis and suggests that increased expression of this enzyme is a physiological response to inflammatory stimuli. Administration of recombinant forms of PAF-AH attenuate inflammation in a variety of experimental models. Conversely, genetic deficiency of PAF-AH in defined human populations increases the severity of atherosclerosis and other syndromes. Recent advances pointing to an interplay among oxidized phospholipid substrates, Lp(a), and PAF-AH could hold the key to a number of unanswered questions.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Inflammation/physiopathology , Oxidative Stress , 1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Animals , Atherosclerosis/physiopathology , Humans , Inflammation Mediators/metabolism , Lipoprotein(a)/metabolism , Severity of Illness Index , Up-Regulation
17.
FASEB J ; 21(4): 1164-76, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17210780

ABSTRACT

Platelet-activating factor acetylhydrolase (PAF-AH) is a phospholipase A2 that inactivates potent lipid messengers, such as PAF and modified phospholipids generated in settings of oxidant stress. The catalytic activity of PAF-AH is sensitive to oxidants, a feature that may have pathological consequences. We report that peroxynitrite, an oxidant species generated after cellular activation, mediates oxidative inactivation of PAF-AH. We found that peroxynitrite inactivated and derivatized the recombinant protein and obtained evidence supporting a role for a methionine and two tyrosine residues in this process. We employed interspecies comparisons and site-directed mutagenesis and identified a role for M-117, and a smaller contribution of Y-307 and Y-335 as targets of oxidant attack using free and lipoprotein-associated recombinant proteins. M-117 is adjacent to W-115 and L-116, which are essential for association of PAF-AH with LDL. Oxidation of LDL-associated PAF-AH partially dissociated the enzyme from the particles. Similarly, oxidation of the purified enzyme in the absence of lipoproteins prevented subsequent association with LDL. These results provide new insights into the molecular mechanisms that mediate inactivation of PAF-AH in settings of oxidant stress and the consequences of oxidation on the ability of this enzyme to associate with LDL.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/blood , 1-Alkyl-2-acetylglycerophosphocholine Esterase/chemistry , Animals , COS Cells , Chlorocebus aethiops , Humans , Lipoproteins/chemistry , Mice , Oxidants/metabolism , Oxidative Stress , Oxygen/metabolism , Peroxynitrous Acid/pharmacology , Phospholipases A/metabolism , Phospholipases A2 , Phospholipids/metabolism , Reactive Oxygen Species , Tyrosine/chemistry
18.
Cell Signal ; 19(9): 1956-63, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17572069

ABSTRACT

Cyclooxygenase-2 is often highly expressed in epithelial malignancies and likely has an active role in tumor development. But how it promotes tumorigenesis is not clearly defined. Recent evidence suggests that this may involve transactivation of the epidermal growth factor receptor through E-prostanoid receptors, but reports differ about the mechanism by which this occurs. We found that E-prostanoid receptors 2-4, but not 1, transactivated the epidermal growth factor receptor. This required metalloproteinase activity, leading to release of growth factors from the cell surface. Both transforming growth factor-alpha and amphiregulin were released in response to over-expression of cyclooxygenase-2, but betacellulin and heparin-binding EGF-like growth factor were not. The metalloproteinase tumor necrosis factor-alpha converting enzyme was required for proteolytic release of transforming growth factor-alpha. We also found that addition of epidermal growth factor receptor ligands to HEK293 cells induced cyclooxygenase-2 expression, suggesting that by activating epidermal growth factor receptor signaling, cyclooxygenase-2 potentially creates a self-perpetuating cycle of cell growth. Consistent with this, inhibition of cyclooxygenase-2 reduced growth of epidermal growth factor receptor over-expressing MCF-10A breast epithelial cells in three-dimensional culture.


Subject(s)
ADAM Proteins/metabolism , Cyclooxygenase 2/metabolism , ErbB Receptors/genetics , Membrane Proteins/metabolism , Receptors, Prostaglandin E/metabolism , Transcriptional Activation/genetics , ADAM17 Protein , Animals , COS Cells , Cell Culture Techniques , Cell Proliferation/drug effects , Chlorocebus aethiops , Cyclooxygenase 2/genetics , Dinoprostone/pharmacology , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Metalloproteases/metabolism , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptional Activation/drug effects , Transforming Growth Factor alpha/metabolism
19.
Article in English | MEDLINE | ID: mdl-17629689

ABSTRACT

Platelet-activating factor (PAF) is a proinflammatory mediator that plays a central role in acute lung injury (ALI). PAF- acetylhydrolases (PAF-AHs) terminate PAF's signals and regulate inflammation. In this study, we describe the kinetics of plasma and bronchoalveolar lavage (BAL) PAF-AH in the early phase of ALI. Six pigs with oleic acid induced ALI and two healthy controls were studied. Plasma and BAL samples were collected every 2h and immunohistochemical analysis of PAF-AH was performed in lung tissues. PAF-AH activity in BAL was increased at the end of the experiment (BAL PAF-AH Time 0=0.001+/-0.001 nmol/ml/min/g vs Time 6=0.031+/-0.018 nmol/ml/min/g, p=0.04) while plasma activity was not altered. We observed increased PAF-AH staining of macrophages and epithelial cells in the lungs of animals with ALI but not in healthy controls. Our data suggest that increases in PAF-AH levels are, in part, a result of alveolar production. PAF-AH may represent a modulatory strategy to counteract the excessive pro-inflammatory effects of PAF and PAF-like lipids in lung inflammation.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/biosynthesis , Lung/enzymology , Respiratory Distress Syndrome/enzymology , 1-Alkyl-2-acetylglycerophosphocholine Esterase/blood , Animals , Bronchoalveolar Lavage Fluid/chemistry , Female , Immunohistochemistry , Kinetics , Lung/metabolism , Oleic Acid , Respiratory Distress Syndrome/chemically induced , Swine , Time Factors
20.
Prostaglandins Other Lipid Mediat ; 82(1-4): 147-54, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17164142

ABSTRACT

The temporal association between loss of function of the tumor suppressor adenomatous polyposis coli (APC) and overexpression of cyclooxygenase 2 (COX-2) has been demonstrated in vivo and has led to the hypothesis that APC regulates COX-2 expression. This could potentially occur through a variety of mechanisms including the well-characterized ability of APC to negatively regulate Wnt signaling and decrease expression of target genes. However, recent findings suggest that the products of COX-2 elicit effects that occur upstream of the beta-catenin/TCF/LEF pathway. This review will focus on the regulation of COX-2 by APC and the interplay between COX-2 and the Wnt signaling pathway.


Subject(s)
Colonic Neoplasms/physiopathology , Cyclooxygenase 2/physiology , Prostaglandins/physiology , Signal Transduction/physiology , Adenomatous Polyposis Coli Protein/physiology , Animals , Dinoprostone/physiology , Gene Expression Regulation, Neoplastic , Genes, APC/physiology , Humans , Up-Regulation , Wnt Proteins/physiology , beta Catenin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL