Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Eur J Pharmacol ; 887: 173559, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32949605

ABSTRACT

There is little literature showing the effect of urocortin (UCN) on macrophage apoptosis. The underlying mechanism is also unclear. This work was to investigate the involvement of UCN in the regulation of LPS-induced macrophage apoptosis and hence in the prevention from the atherosclerotic lesion development through targeting PLA2. Flow cytometry analysis showed that cell apoptosis was increased by more than 50% after LPS treatment in human THP-1 macrophage. Lp-PLA2 and cPLA2 were found to mediate LPS-induced macrophage apoptosis and NF-κB differentially influenced the expression of Lp-PLA2 and cPLA2. However, the reverse regulation of the expression of Lp-PLA2 and cPLA2 by NF-κB suggested that NF-κB may not be a key target for regulating macrophage apoptosis. Interestingly, we found that the approximate three folds upregulation of cPLA2 was in line with the induction of S1P formation and cell apoptosis by LPS. Inversely, LPS obviously decreased UCN expression by about 50% and secretion by about 25%. Both the enzyme inhibitor and knockdown expression of cPLA2 could completely abolish LPS-induced cell apoptosis. In addition, suppression of S1P synthesis by Sphk1 inhibitor PF-543 reduced the expression of cPLA2 and cell apoptosis but at the same time restored the normal level of UCN in cell culture supernatant. Furthermore, addition of exogenous UCN also reversed LPS-induced expression of cPLA2 and apoptosis. Taken together, UCN may be the reverse regulator of LPS-S1P-cPLA2-apoptosis pathway, thereby contributing to the prevention from the formation of unstable plaques.


Subject(s)
Apoptosis/drug effects , Lipopolysaccharides/pharmacology , Phospholipases A2, Cytosolic/drug effects , Proprotein Convertases/drug effects , Serine Endopeptidases/drug effects , Signal Transduction/drug effects , THP-1 Cells/drug effects , Urocortins/physiology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Atherosclerosis/drug therapy , Atherosclerosis/pathology , Gene Expression Regulation/drug effects , Humans , Methanol/pharmacology , Mitogen-Activated Protein Kinases/pharmacology , NF-kappa B/drug effects , Phospholipases A2, Cytosolic/biosynthesis , Pyrrolidines/pharmacology , Sulfones/pharmacology , Urocortins/pharmacology
2.
Neurochem Int ; 129: 104497, 2019 10.
Article in English | MEDLINE | ID: mdl-31251945

ABSTRACT

Chronic alcoholism promotes brain damage that impairs memory and cognition. High binge alcohol levels in adult rats also cause substantial neurodamage to memory-linked regions, notably, the hippocampus (HC) and entorhinal cortex (ECX). Concurrent with neurodegeneration, alcohol elevates poly (ADP-ribose) polymerase-1 (PARP-1) and cytosolic phospholipase A2 (cPLA2) levels. PARP-1 triggers necrosis when excessively activated, while cPLA2 liberates neuroinflammatory ω-6 arachidonic acid. Inhibitors of PARP exert in vitro neuroprotection while suppressing cPLA2 elevations in alcohol-treated HC-ECX slice cultures. Here, we examined in vivo neuroprotection and cPLA2 suppression by the PARP inhibitor, veliparib, in a recognized adult rat model of alcohol-binging. Adult male rats received Vanilla Ensure containing alcohol (ethanol, 7.1 ±â€¯0.3 g/kg/day), or control (dextrose) ±â€¯veliparib (25 mg/kg/day), by gavage 3x daily for 4 days. Rats were sacrificed on the morning after the final binge. HC and ECX neurodegeneration was assessed in fixed sections by Fluoro-Jade B (FJB) staining. Dorsal HC, ventral HC, and ECX cPLA2 levels were quantified by immunoblotting. Like other studies using this model, alcohol binges elevated FJB staining in the HC (dentate gyrus) and ECX, indicating neurodegeneration. Veliparib co-treatment significantly reduced dentate gyrus and ECX neurodegeneration by 79% and 66%, respectively. Alcohol binges increased cPLA2 in the ventral HC by 34% and ECX by 72%, which veliparib co-treatment largely prevented. Dorsal HC cPLA2 levels remained unaffected by alcohol binges, consistent with negligible FJB staining in this brain region. These in vivo results support an emerging key role for PARP in binge alcohol-induced neurodegeneration and cPLA2-related neuroinflammation.


Subject(s)
Alcohol-Induced Disorders, Nervous System/prevention & control , Benzimidazoles/therapeutic use , Nerve Tissue Proteins/biosynthesis , Phospholipases A2, Cytosolic/biosynthesis , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Alcohol-Induced Disorders, Nervous System/drug therapy , Alcohol-Induced Disorders, Nervous System/enzymology , Animals , Benzimidazoles/pharmacology , Binge Drinking , Dentate Gyrus/drug effects , Dentate Gyrus/enzymology , Dentate Gyrus/pathology , Disease Models, Animal , Entorhinal Cortex/drug effects , Entorhinal Cortex/enzymology , Entorhinal Cortex/pathology , Enzyme Induction/drug effects , Male , Nerve Tissue Proteins/genetics , Phospholipases A2, Cytosolic/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley
3.
Vascul Pharmacol ; 71: 116-26, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25869501

ABSTRACT

This work is to investigate the role of CRF receptors in VSMC migration and the relevant mechanisms. We studied the role of CRF receptors in cell motility and found that S1P signaling pathway is involved in the regulation of cPLA2 induced by CRF. S1P is synthesized by Sphk1 and Sphk2 and binds to five GPCR designated S1P1-5. We observed that activation of CRFR1 resulted in increased cell migration, whereas activation of CRFR2 resulted in decreased cell migration. cPLA2 and iPLA2 were knocked down respectively to explore the corresponding effect on cell migration by means of shRNA interference. cPLA2 expression was increased by CRFR1 but decreased by CRFR2. On the contrary, iPLA2 expression was inhibited by CRFR1 but enhanced by CRFR2. The regulation of cPLA2 was in line with the regulation of Sphk1 and hence cell migration after the activation of CRFR1 or CRFR2. Consistently, S1P release was enhanced with CRFR1 activation. Both DMS (Sphk inhibitor) and CAY10444 (S1PR3 inhibitor) attenuated cPLA2 expression and thus decreased cell migration in response to CRF. In addition, CRF could not promote cell migration after S1PR3 silencing. Our results suggest the pro-migratory role of CRFR1-Sphk1-S1P-S1PR3-cPLA2 signaling pathway in VSMCs.


Subject(s)
Cell Movement/physiology , Corticotropin-Releasing Hormone/pharmacology , Muscle, Smooth, Vascular/metabolism , Phospholipases A2, Cytosolic/biosynthesis , Receptors, Lysosphingolipid/biosynthesis , Animals , Cell Line , Cell Movement/drug effects , Cells, Cultured , Gene Expression Regulation, Enzymologic , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/metabolism , Sphingosine-1-Phosphate Receptors
4.
Mol Neurobiol ; 51(3): 1103-15, 2015.
Article in English | MEDLINE | ID: mdl-24915969

ABSTRACT

Bradykinin (BK) is a proinflammatory mediator and elevated in several brain injury and inflammatory diseases. The deleterious effects of BK on brain astrocytes may aggravate brain inflammation mediated through the upregulation of cytosolic phospholipase A2 (cPLA2)/cyclooxygenase-2 (COX-2)-derived prostaglandin E2 (PGE2) production. However, the signaling mechanisms underlying BK-induced cPLA2 expression in brain astrocytes remain unclear. Herein, we investigated the effects of activation of cPLA2/COX-2 system on BK-induced cPLA2 upregulation in rat brain astrocytes (RBA-1). The data obtained with Western blotting, RT-PCR, and immunofluorescent staining analyses showed that BK-induced de novo cPLA2 expression was mediated through activation of cPLA2/COX-2 system. Upregulation of native cPLA2/COX-2 system by BK through activation of PKCδ, c-Src, MAPKs (ERK1/2 and JNK1/2) cascades led to PGE2 biosynthesis and release. Subsequently, the released PGE2 induced cPLA2 expression via the same signaling pathways (PKCδ, c-Src, ERK1/2, and JNK1/2) and then activated the cyclic AMP response element-binding protein (CREB) via B2 BK receptor-mediated cPLA2/COX-2 system-derived PGE2/EP-dependent manner. Finally, upregulation of cPLA2 by BK may promote more PGE2 production. These results demonstrated that in RBA-1, activation of CREB by PGE2/EP-mediated PKCδ/c-Src/MAPK cascades is essential for BK-induced de novo cPLA2 protein. More importantly, upregulation of cPLA2 by BK through native cPLA2/COX-2 system may be a positive feedback mechanism that enhances prolonged brain inflammatory responses. Understanding the mechanisms of cPLA2/COX-2 system upregulated by BK on brain astrocytes may provide rational therapeutic interventions for brain injury and inflammatory diseases.


Subject(s)
Astrocytes/metabolism , Bradykinin/pharmacology , Brain/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Phospholipases A2, Cytosolic/biosynthesis , Animals , Astrocytes/drug effects , Autocrine Communication/drug effects , Autocrine Communication/physiology , Brain/drug effects , Gene Expression Regulation, Enzymologic , Male , Rats
5.
Exp Biol Med (Maywood) ; 237(5): 499-508, 2012 May.
Article in English | MEDLINE | ID: mdl-22442340

ABSTRACT

PG201, an ethanol extract from a mixture of 12 herbs, has strong antiarthritic activity. To understand the molecular mechanisms underlying its anti-inflammatory effects, PG201-mediated suppression of inflammatory mediators was studied in Raw264.7, a mouse macrophage cell line. PG201 decreased the expression of interleukin (IL)-1ß, IL-6 and CC chemokine ligand-2, but not tumor necrosis factor-α, at the protein and mRNA levels in lipopolysaccharide-stimulated Raw264.7 cells. Results from a gel retardation assay indicated that PG201 substantially reduced the DNA-binding activity of the activator protein-1 and cyclic adenosine monophosphate-responsive element-binding protein transcription factors, but not nuclear factor-κB. Western blot and Northern blot analyses showed that PG201 reduced inducible nitric oxide synthase and cytosolic phospholipase A(2) (cPLA(2)) protein expression, but did not affect mRNA expression, ultimately resulting in decreased nitric oxide and prostaglandin E(2). The protein expression of cPLA(2) was decreased by PG201 in the presence of cycloheximide, an inhibitor of translation, suggesting that PG201 may facilitate the degradation of cPLA(2). Taken together, these results suggest that PG201 selectively affects the expression of proteins that play key roles in the inflammatory response at transcriptional and post-translational levels.


Subject(s)
Arthritis, Rheumatoid/immunology , Inflammation Mediators/metabolism , Macrophages/drug effects , Macrophages/immunology , Osteoarthritis/immunology , Plant Extracts/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Arthritis, Rheumatoid/pathology , Cell Line , Chemokine CCL2/biosynthesis , Cycloheximide/pharmacology , Dinoprostone/analysis , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Lipopolysaccharides/immunology , Macrophages/metabolism , Mice , NF-kappa B/biosynthesis , Nitric Oxide/analysis , Nitric Oxide Synthase Type II/biosynthesis , Osteoarthritis/pathology , Phospholipases A2, Cytosolic/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factor AP-1/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis
6.
Neurobiol Aging ; 33(10): 2422-30, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22226487

ABSTRACT

During heat waves many elderly individuals die as a consequence of dehydration. This is partially due to deficits in mechanisms controlling thirst. Reduced thirst following dipsogenic stimuli is well documented in aged humans and rodents. Low in vivo long-chain omega-3 fatty acid levels, as can occur in aging, have been shown to alter body fluid and sodium homeostasis. Therefore, the effect of dietary omega-3 fatty acid supplementation on drinking responses in aged rats was examined. Omega-3 fatty acids reversed thirst deficits in aged rats following dehydration and hypertonic stimuli; angiotensin (ANG) II induced drinking was unaffected in aged rats. Plasma atrial natriuretic peptide (ANP) and arginine vasopressin (AVP) were altered with age, but not affected by diet. Aged omega-3 fatty acid deficient animals displayed increased hypothalamic cytosolic phospholipase A(2) (cPLA(2)), cyclooxygenase (COX)-2, and prostaglandin E (PGE) synthase messenger (m)RNA expression, compared with animals that received omega-3 fatty acids. The aged low omega-3 fatty acid fed animals had significantly elevated hypothalamic PGE(2) compared with all other groups. Hypothalamic PGE(2) was negatively correlated with drinking induced by both dehydration and hypertonicity. The results indicate that PGE(2) may be the underlying mechanism of the reduced thirst observed in aging.


Subject(s)
Dietary Supplements , Fatty Acids, Omega-3/administration & dosage , Thirst/drug effects , Age Factors , Angiotensin II/pharmacology , Animals , Arginine Vasopressin/blood , Atrial Natriuretic Factor/blood , Cyclooxygenase 2/biosynthesis , Dehydration/drug therapy , Dehydration/physiopathology , Dinoprostone/analysis , Drinking/drug effects , Drinking/physiology , Hypothalamus/chemistry , Hypothalamus/enzymology , Intramolecular Oxidoreductases/biosynthesis , Male , Phospholipases A2, Cytosolic/biosynthesis , Prostaglandin-E Synthases , Rats , Saline Solution, Hypertonic/pharmacology , Thirst/physiology
7.
Behav Brain Res ; 226(2): 386-96, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21971013

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder, characterized by hypokinesia, but also mood and cognitive disorders. Neuropathologically, PD involves loss of nigrostriatal dopamine (DA) and secondary non-dopaminergic abnormalities. Inflammation may contribute to PD pathogenesis, evident by increased production of pro-inflammatory cytokines. PD onset has been positively associated with dietary intake of omega-(n)-6 polyunsaturated fatty acids (PUFA). On the other hand, omega-(n)-3 PUFA may benefit PD. One of these n-3 PUFA, eicosapentaenoic acid (EPA), is a neuroprotective lipid with anti-inflammatory properties, but its neuroprotective effects in PD are unknown. Thus, we presently tested the hypothesis that EPA can protect against behavioral impairments, neurodegeneration and inflammation in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-probenecid (MPTP-P) mouse model of PD. MPTP-P injections caused hypokinesia in the rotorod and pole test, hyperactivity in the open field, and impaired mice on the cued version (procedural memory) of the Morris water maze. MPTP-P caused a loss of nigrostriatal DA and altered neurochemistry in the frontal cortex and hippocampus. Furthermore, striatal levels of pro-inflammatory cytokines were increased, while the brain n-3/n-6 lipid profile remained unaltered. Feeding mice a 0.8% ethyl-eicosapentaenoate (E-EPA) diet prior to MPTP-P injections increased brain EPA and docosapentaenoic acid (DPA) but not docosahexaenoic acid (DHA) or n-6 PUFA. The diet attenuated the hypokinesia induced by MPTP-P and ameliorated the procedural memory deficit. E-EPA also suppressed the production of pro-inflammatory cytokines. However, E-EPA did not prevent nigrostriatal DA loss. Based on this partial protective effect of E-EPA, further testing may be warranted.


Subject(s)
Eicosapentaenoic Acid/analogs & derivatives , Inflammation/diet therapy , MPTP Poisoning/diet therapy , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/diet therapy , Rotarod Performance Test/statistics & numerical data , Animals , Biogenic Monoamines/metabolism , Brain/drug effects , Brain/enzymology , Brain/metabolism , Cyclooxygenase 2/biosynthesis , Cytokines/metabolism , Eicosapentaenoic Acid/pharmacology , Eicosapentaenoic Acid/therapeutic use , Exploratory Behavior/drug effects , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-6/metabolism , Humans , Inflammation/chemically induced , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Neuroprotective Agents/therapeutic use , Parkinsonian Disorders/chemically induced , Phospholipases A2, Cytosolic/biosynthesis , Probenecid
8.
Basic Res Cardiol ; 106(3): 431-46, 2011 May.
Article in English | MEDLINE | ID: mdl-21360043

ABSTRACT

Pioglitazone (PIO), a PPAR-γ agonist, limits myocardial infarct size by activating Akt and upregulating cytosolic phospholipase A(2) (cPLA(2)) and cyclooxygenase (COX)-2. However, PIO has several PPAR-γ-independent effects. We assessed whether PIO limits myocardial infarct size in PPAR-γ-knockout mice, attenuates hypoxia-reoxygenation injury and upregulates P-Akt, cPLA(2), and COX-2 expression in PPAR-γ-knockout cardiomyocytes. Cardiac-specific inducible PPAR-γ knockout mice were generated by crossing αMHC-Cre mice to PPAR-γ(loxp/loxp) mice. PPAR-γ deletion was achieved after 7 days of intraperitoneal tamoxifen (20 mg/kg/day) administration. Mice received PIO (10 mg/kg/day), or vehicle, for 3 days and underwent coronary occlusion (30 min) followed by reperfusion (4 h). We assessed the area at risk by blue dye and infarct size by TTC. Cultured adult cardiomyocytes of PPAR-γ(loxp/loxp/cre) mice without or with pretreatment with tamoxifen were incubated with or without PIO and subjected to 2 h hypoxia/2 h reoxygenation. Cardiac-specific PPAR-γ knockout significantly increased infarct size. PIO reduced infarct size by 51% in PPAR-γ knockout mice and by 55% in mice with intact PPAR-γ. Deleting the PPAR-γ gene increased cell death in vitro. PIO reduced cell death in cells with and without intact PPAR-γ. PIO similarly increased myocardial Ser-473 P-Akt, cPLA(2), and COX-2 levels after hypoxia/reoxygenation in cells with and without intact PPAR-γ. PIO limited infarct size in mice in a PPAR-γ-independent manner. PIO activated Akt, increased the expression of cPLA(2) and COX-2, and protected adult cardiomyocytes against the effects of hypoxia/reoxygenation independent of PPAR-γ activation.


Subject(s)
Cyclooxygenase 2/biosynthesis , Hypoglycemic Agents/pharmacology , Myocardial Infarction/prevention & control , Phospholipases A2, Cytosolic/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Thiazolidinediones/pharmacology , Animals , Enzyme Activation/drug effects , Gene Expression , Immunoblotting , Immunohistochemistry , Mice , Mice, Knockout , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism , PPAR gamma/metabolism , Pioglitazone , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Up-Regulation
9.
J Cell Biochem ; 109(5): 1045-56, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20069553

ABSTRACT

Cytosolic phospholipase A(2) (cPLA(2)) plays a pivotal role in mediating agonist-induced arachidonic acid (AA) release for prostaglandin (PG) synthesis during inflammation triggered by IL-1beta. However, the mechanisms underlying IL-1beta-induced cPLA(2) expression and PGE(2) synthesis in human tracheal smooth muscle cells (HTSMCs) remain unknown. IL-1beta-induced cPLA(2) protein and mRNA expression, PGE(2) production, or phosphorylation of p42/p44 MAPK, p38 MAPK, and JNK1/2, which was attenuated by pretreatment with the inhibitors of MEK1/2 (U0126), p38 MAPK (SB202190), and JNK1/2 (SP600125) or transfection with siRNAs of MEK1, p42, p38, and JNK2. IL-1beta-induced cPLA(2) expression was also inhibited by pretreatment with a NF-kappaB inhibitor, helenalin or transfection with siRNA of NIK, IKKalpha, or IKKbeta. IL-beta-induced NF-kappaB translocation was blocked by pretreatment with helenalin, but not U0126, SB202190, and SP600125. In addition, transfection with p300 siRNA blocked cPLA(2) expression induced by IL-1beta. Moreover, p300 was associated with the cPLA(2) promoter, which was dynamically linked to histone H4 acetylation stimulated by IL-1beta. These results suggest that in HTSMCs, activation of MAPKs, NF-kappaB, and p300 are essential for IL-1beta-induced cPLA(2) expression and PGE(2) secretion.


Subject(s)
E1A-Associated p300 Protein/metabolism , Interleukin-1beta/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Myocytes, Smooth Muscle/enzymology , NF-kappa B/metabolism , Phospholipases A2, Cytosolic/biosynthesis , Trachea/cytology , Dinoprostone/metabolism , Enzyme Activation/drug effects , Enzyme Induction/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Smooth Muscle/drug effects , Phospholipases A2, Cytosolic/genetics , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Biosynthesis/drug effects , Proto-Oncogene Proteins/metabolism , Transcription, Genetic/drug effects
10.
Mol Psychiatry ; 15(6): 602-14, 2010 Jun.
Article in English | MEDLINE | ID: mdl-18982003

ABSTRACT

Agents effective against mania in bipolar disorder are reported to decrease turnover of arachidonic acid (AA) in phospholipids and expression of calcium-dependent AA-selective cytosolic phospholipase A(2) (cPLA(2)) in rat brain. In contrast, fluoxetine, an antidepressant that is reported to switch bipolar depressed patients to mania, increases cPLA(2) expression and AA turnover in rat brain. We therefore hypothesized that antidepressants that increase switching to mania generally increase cPLA(2) and AA turnover in brain. To test this hypothesis, adult male CDF-344 rats were administered imipramine and bupropion, with reported high and low switching rates, respectively, at daily doses of 10 and 30 mg kg(-1) i.p., respectively, or i.p. saline (control) for 21 days. Frontal cortex expression of different PLA(2) enzymes and AA turnover rates in brain when the rats were unanesthetized were measured. Compared with chronic saline, chronic imipramine but not bupropion significantly increased cortex cPLA(2) mRNA activity, protein and phosphorylation, expression of the cPLA(2) transcription factor, activator protein-2alpha (AP-2alpha) and AA turnover in phospholipids. Protein levels of secretory phospholipase A(2), calcium-independent phospholipase A(2), cyclooxygenase (COX)-1 and COX-2 were unchanged, and prostaglandin E(2) was unaffected. These results, taken with prior data on chronic fluoxetine in rats, suggest that antidepressants that increase the switching tendency of bipolar depressed patients to mania do so by increasing AA recycling and metabolism in brain. Mania in bipolar disorder thus may involve upregulated brain AA metabolism.


Subject(s)
Arachidonic Acid/metabolism , Bipolar Disorder/diagnosis , Bipolar Disorder/metabolism , Bupropion/pharmacology , Frontal Lobe/drug effects , Imipramine/pharmacology , Signal Transduction/drug effects , Animals , Bupropion/administration & dosage , Cyclooxygenase 1/biosynthesis , Cyclooxygenase 2/biosynthesis , Dinoprostone/biosynthesis , Drug Administration Schedule , Frontal Lobe/metabolism , Humans , Imipramine/administration & dosage , Male , Phospholipases A2, Calcium-Independent/biosynthesis , Phospholipases A2, Cytosolic/biosynthesis , Phosphorylation/drug effects , Rats , Transcription Factor AP-2/biosynthesis , Up-Regulation/drug effects
11.
Eur J Haematol ; 79(6): 468-76, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17976189

ABSTRACT

Leukotrienes (LT) exert stimulatory effects on myelopoiesis, beside their inflammatory and immunomodulating effects. Here, we have studied the expression and activity of the enzymes involved in the synthesis of leukotriene B4 (LTB4) in acute myeloid leukemia (AML) cells (16 clones) and G-CSF mobilized peripheral blood CD34+ cells. CD34+ cells from patients with non-myeloid malignancies expressed cytosolic phospholipase A2 (cPLA2), 5-lipoxygenase activating protein (FLAP), and leukotriene A4 (LTA4) hydrolase but not 5-lipoxygenase (5-LO). The enzyme cPLA2 was abundantly expressed in AML cells and the activity of the enzyme was high in certain AML clones. The expression of 5-LO, FLAP, and LTA4 hydrolase in AML clones was in general lower than in healthy donor polymorphonuclear leukocytes (PMNL). The calcium ionophore A23187-induced release of [14C] arachidonic acid (AA) in AML cells was low, compared with PMNL, and did not correlate with the expression of cPLA2 protein. Biosynthesis of LTB4, upon calcium ionophore A23187 activation, was only observed in five of the investigated AML clones and only three of the most differentiated clones produced similar amounts of LTB4 as PMNL. The capacity of various cell clones to produce LTs could neither be explained by the difference in [1-(14)C] AA release nor 5-LO expression. Taken together, these results indicate that LT synthesis is under development during early myelopoiesis and the capacity to produce LTs is gained upon maturation. High expression of cPLA2 in AML suggests a putative role of this enzyme in the pathophysiology of this disease.


Subject(s)
Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/blood , Leukotriene B4/biosynthesis , Phospholipases A2, Cytosolic/biosynthesis , Adult , Aged , Antigens, CD34/biosynthesis , Arachidonate 5-Lipoxygenase/biosynthesis , Calcimycin/pharmacology , Female , Granulocyte Colony-Stimulating Factor/metabolism , Humans , Ionophores/pharmacology , Male , Middle Aged , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL
...