Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Lipids ; 54(9): 557-570, 2019 09.
Article in English | MEDLINE | ID: mdl-31475368

ABSTRACT

The remodeling of membrane lipids is a mechanism that allows microorganisms to survive in unfavorable environments such as industrial effluents, which often contain inorganic and organic pollutants, like chromium and phenol. In the present work, we evaluated the effect of Cr(VI) and phenol on the membrane of Acinetobacter guillouiae SFC 500-1A, a bacterial strain isolated from tannery sediments where such pollutants can be found. The presence of lipid kinases and phospholipases and the changes in their activities under exposure to these pollutants were determined. Cr(VI) and Cr(VI) + phenol caused the membrane to become more rigid for up to 16 h after exposure. This could be due to an increase in cardiolipin (Ptd2 Gro) and a decrease in phosphatidylethanolamine (PtdEtn), which are indicative of more order and rigidity in the membrane. Increased phospholipase A activity (PLA, EC 3.1.1.4) could be responsible for the decrease in PtdEtn levels. Moreover, our results indicate that Cr(VI) and Cr(VI) + phenol trigger the phosphatidic acid (PtdOH) signal. The finding of significantly increased phosphatidylinositol-4-phosphate (PtdIns-4-P) levels means this is likely achieved via PtdIns-PLC/DGK. This report provides the first evidence that A. guillouiae SFC 500-1A is able to sense Cr(VI) and phenol, transduce this signal through changes in the physical state of the membrane, and trigger lipid-signaling events.


Subject(s)
Acinetobacter/drug effects , Cell Membrane/drug effects , Chromium/pharmacology , Phenols/pharmacology , Phosphatidic Acids/antagonists & inhibitors , Signal Transduction/drug effects , Cell Membrane/metabolism , Fluorescence Polarization , Phosphatidic Acids/metabolism
2.
J Biol Chem ; 294(46): 17168-17185, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31515268

ABSTRACT

The homeostasis of most organelles requires membrane fusion mediated by soluble N -ethylmaleimide-sensitive factor (NSF) attachment protein receptors (SNAREs). SNAREs undergo cycles of activation and deactivation as membranes move through the fusion cycle. At the top of the cycle, inactive cis-SNARE complexes on a single membrane are activated, or primed, by the hexameric ATPase associated with the diverse cellular activities (AAA+) protein, N-ethylmaleimide-sensitive factor (NSF/Sec18), and its co-chaperone α-SNAP/Sec17. Sec18-mediated ATP hydrolysis drives the mechanical disassembly of SNAREs into individual coils, permitting a new cycle of fusion. Previously, we found that Sec18 monomers are sequestered away from SNAREs by binding phosphatidic acid (PA). Sec18 is released from the membrane when PA is hydrolyzed to diacylglycerol by the PA phosphatase Pah1. Although PA can inhibit SNARE priming, it binds other proteins and thus cannot be used as a specific tool to further probe Sec18 activity. Here, we report the discovery of a small-molecule compound, we call IPA (inhibitor of priming activity), that binds Sec18 with high affinity and blocks SNARE activation. We observed that IPA blocks SNARE priming and competes for PA binding to Sec18. Molecular dynamics simulations revealed that IPA induces a more rigid NSF/Sec18 conformation, which potentially disables the flexibility required for Sec18 to bind to PA or to activate SNAREs. We also show that IPA more potently and specifically inhibits NSF/Sec18 activity than does N-ethylmaleimide, requiring the administration of only low micromolar concentrations of IPA, demonstrating that this compound could help to further elucidate SNARE-priming dynamics.


Subject(s)
Adenosine Triphosphatases/genetics , Ethylmaleimide/metabolism , Phosphatidic Acids/chemistry , Saccharomyces cerevisiae Proteins/genetics , Small Molecule Libraries/chemistry , Vesicular Transport Proteins/genetics , ATPases Associated with Diverse Cellular Activities/chemistry , ATPases Associated with Diverse Cellular Activities/genetics , Adenosine Triphosphatases/chemistry , Membrane Fusion/drug effects , Membrane Fusion/genetics , Membrane Lipids/chemistry , Membrane Lipids/genetics , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Molecular Dynamics Simulation , N-Ethylmaleimide-Sensitive Proteins/chemistry , N-Ethylmaleimide-Sensitive Proteins/genetics , Phosphatidic Acids/antagonists & inhibitors , SNARE Proteins/chemistry , SNARE Proteins/genetics , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Small Molecule Libraries/pharmacology , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/chemistry , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/genetics , Vacuoles/genetics , Vesicular Transport Proteins/chemistry
3.
J Pain ; 14(1): 66-78, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23273834

ABSTRACT

UNLABELLED: Growing evidence indicates that various chronic pain syndromes exhibit tissue abnormalities caused by microvasculature dysfunction in the blood vessels of skin, muscle, or nerve. We tested whether topical combinations aimed at improving microvascular function would relieve allodynia in animal models of complex regional pain syndrome type I (CRPS-I) and neuropathic pain. We hypothesized that topical administration of either α(2)-adrenergic (α(2)A) receptor agonists or nitric oxide (NO) donors combined with either phosphodiesterase (PDE) or phosphatidic acid (PA) inhibitors would effectively reduce allodynia in these animal models of chronic pain. Single topical agents produced significant dose-dependent antiallodynic effects in rats with chronic postischemia pain, and the antiallodynic dose-response curves of PDE and PA inhibitors were shifted 2.5- to 10-fold leftward when combined with nonanalgesic doses of α(2)A receptor agonists or NO donors. Topical combinations also produced significant antiallodynic effects in rats with sciatic nerve injury, painful diabetic neuropathy, and chemotherapy-induced painful neuropathy. These effects were shown to be produced by a local action, lasted up to 6 hours after acute treatment, and did not produce tolerance over 15 days of chronic daily dosing. The present results support the hypothesis that allodynia in animal models of CRPS-I and neuropathic pain is effectively relieved by topical combinations of α(2)A or NO donors with PDE or PA inhibitors. This suggests that topical treatments aimed at improving microvascular function may reduce allodynia in patients with CRPS-I and neuropathic pain. PERSPECTIVE: This article presents the synergistic antiallodynic effects of combinations of α(2)A or NO donors with PDE or PA inhibitors in animal models of CRPS-I and neuropathic pain. The data suggest that effective clinical treatment of chronic neuropathic pain may be achieved by therapies that alleviate microvascular dysfunction in affected areas.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/therapeutic use , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Microcirculation/drug effects , Neuralgia/drug therapy , Neuralgia/physiopathology , Nitric Oxide Donors/therapeutic use , Phosphatidic Acids/antagonists & inhibitors , Phosphodiesterase Inhibitors/therapeutic use , Reflex Sympathetic Dystrophy/drug therapy , Reflex Sympathetic Dystrophy/physiopathology , Administration, Topical , Adrenergic alpha-2 Receptor Agonists/administration & dosage , Animals , Chemistry, Pharmaceutical , Diabetes Mellitus, Experimental/complications , Diabetic Neuropathies/drug therapy , Drug Combinations , Male , Nitric Oxide Donors/administration & dosage , Ointments , Oxygen Consumption , Pain Measurement/drug effects , Phosphodiesterase Inhibitors/administration & dosage , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Regional Blood Flow/drug effects , Sciatic Neuropathy/drug therapy
4.
Dev Biol ; 362(2): 154-61, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22178154

ABSTRACT

In order to fertilize the oocyte, sperm must undergo a series of biochemical changes in the female reproductive tract, known as capacitation. Once capacitated, spermatozoon can bind to the zona pellucida of the egg and undergo the acrosome reaction (AR), a process that enables its penetration and fertilization of the oocyte. Important processes that characterize sperm capacitation are actin polymerization and the development of hyper-activated motility (HAM). Previously, we showed that Phospholipase D (PLD)-dependent actin polymerization occurs during sperm capacitation, however the role of this process in sperm capacitation is not yet known. In the present study, we showed for the first time the involvement of PLD-dependent actin polymerization in sperm motility during mouse and human capacitation. Sperm incubated under capacitation conditions revealed a time dependent increase in actin polymerization and HAM. Inhibition of Phosphatidic Acid (PA) formation by PLD using butan-1-ol, inhibited actin polymerization and motility, as well as in vitro fertilization (IVF) and the ability of the sperm to undergo the AR. The inhibition of sperm HAM by low concentration of butan-1-ol is completely restored by adding PA, further indicating the involvement of PLD in these processes. Furthermore, exogenous PA enhanced rapid actin polymerization that was followed by a rise in the HAM, as well as an increased in IVF rate. In conclusion, our results demonstrate that PLD-dependent actin polymerization is a critical step needed for the development of HAM during mouse and human sperm capacitation.


Subject(s)
Acrosome Reaction/physiology , Actins/metabolism , Phospholipase D/metabolism , Sperm Capacitation/physiology , Sperm Motility/physiology , Zona Pellucida/metabolism , 1-Butanol/pharmacology , Animals , Female , Fertilization in Vitro , Humans , Immunoblotting , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Phosphatidic Acids/antagonists & inhibitors , Phosphatidic Acids/metabolism , Polymerization/drug effects , Sperm Motility/drug effects
5.
J Immunol ; 183(8): 5104-12, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19794068

ABSTRACT

Initial IgE-dependent signaling events are associated with detergent-resistant membrane microdomains. Following Ag stimulation, the IgE-receptor (Fc(epsilon)RI ) accumulates within these domains. This facilitates the phosphorylation of Fc(epsilon)RI subunits by the Src kinase, Lyn, and the interaction with adaptor proteins, such as the linker for activation of T cells. Among the phospholipases (PL) subsequently activated, PLD is of interest because of its presence in lipid microdomains and the possibility that its product, phosphatidic acid, may regulate signal transduction and membrane trafficking. We find that in Ag-stimulated RBL-2H3 mast cells, the association of Fc(epsilon)RI with detergent-resistant membrane fractions is inhibited by 1-butanol, which subverts production of phosphatidic acid to the biologically inert phosphatidylbutanol. Furthermore, the knockdown of PLD2, and to a lesser extent PLD1 with small inhibitory RNAs, also suppressed the accumulation of Fc(epsilon)RI and Lyn in these fractions as well as the phosphorylation of Src kinases, Fc(epsilon)RI , linker for activation of T cells, and degranulation. These effects were accompanied by changes in distribution of the lipid microdomain component, ganglioside 1, in the plasma membrane as determined by binding of fluorescent-tagged cholera toxin B subunit and confocal microscopy in live cells. Collectively, these findings suggest that PLD activity plays an important role in promoting IgE-dependent signaling events within lipid microdomains in mast cells.


Subject(s)
Mast Cells/enzymology , Membrane Microdomains/immunology , Phospholipase D/metabolism , Receptors, IgE/metabolism , 1-Butanol/pharmacology , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Degranulation/immunology , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/enzymology , Dinitrophenols/immunology , Gene Knockdown Techniques , Glycerophospholipids/immunology , Glycerophospholipids/metabolism , Mast Cells/immunology , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Membrane Proteins/immunology , Membrane Proteins/metabolism , Phosphatidic Acids/antagonists & inhibitors , Phosphatidic Acids/immunology , Phosphatidic Acids/metabolism , Phospholipase D/genetics , Phosphoproteins/immunology , Phosphoproteins/metabolism , Phosphorylation/immunology , RNA, Small Interfering/immunology , RNA, Small Interfering/metabolism , Rats , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Receptors, IgE/drug effects , Serum Albumin, Bovine/immunology , Signal Transduction/immunology , Thy-1 Antigens/immunology , Thy-1 Antigens/metabolism , Transfection , beta-Cyclodextrins/pharmacology , src-Family Kinases/immunology , src-Family Kinases/metabolism
6.
J Inorg Biochem ; 103(11): 1497-503, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19740543

ABSTRACT

In acid soils, aluminium (Al) toxicity and phosphate (Pi) deficiency are the most significant constraints on plant growth. Al inhibits cell growth and disrupts signal transduction processes, thus interfering with metabolism of phospholipase C (PLC), an enzyme involved in second messenger production in the cell. Using a Coffea arabica suspension cell model, we demonstrate that cell growth inhibition by Al toxicity is mitigated at a high Pi concentration. Aluminium-induced cell growth inhibition may be due to culture medium Pi deficiency, since Pi forms complexes with Al, reducing Pi availability to cells. Phosphate does not mitigate inhibition of PLC activity by Al toxicity. Other enzymes of the phosphoinositide signal transduction pathway were also evaluated. Aluminium disrupts production of second messengers such as inositol 1,4,5-trisphosphate (IP(3)) and phosphatidic acid (PA) by blocking PLC activity; however, phospholipase D (PLD) and diacylglycerol kinase (DGK) activities are stimulated by Al, a response probably aimed at counteracting Al effects on PA formation. Phosphate deprivation also induces PLC and DGK activity. These results suggest that Al-induced cell growth inhibition is not linked to PLC activity inhibition.


Subject(s)
Aluminum/pharmacology , Coffea/drug effects , Phosphates/pharmacology , Signal Transduction/drug effects , Type C Phospholipases/metabolism , Aluminum/analysis , Cells, Cultured , Coffea/growth & development , Coffea/metabolism , Diacylglycerol Kinase/drug effects , Diacylglycerol Kinase/metabolism , Inositol 1,4,5-Trisphosphate/antagonists & inhibitors , Inositol 1,4,5-Trisphosphate/metabolism , Phosphatidic Acids/antagonists & inhibitors , Phosphatidic Acids/metabolism , Phospholipase D/drug effects , Phospholipase D/metabolism , Signal Transduction/physiology , Type C Phospholipases/antagonists & inhibitors
7.
Biochem Biophys Res Commun ; 370(1): 169-73, 2008 May 23.
Article in English | MEDLINE | ID: mdl-18358832

ABSTRACT

While a mother's excessive alcohol consumption during pregnancy is known to have adverse effects on fetal neural development, little is known about the underlying mechanism of these effects. In order to investigate these mechanisms, we investigated the toxic effect of ethanol (ETOH) on neural stem/precursor cell (NSC) proliferation. In cultures of NSCs, phospholipase D (PLD) is activated following stimulation with epidermal growth factor (EGF) and fibroblast growth factor 2 (FGF2). Exposure of NSCs to ETOH suppresses cell proliferation, while it has no effect on cell death. Phosphatidic acid (PA), which is a signaling messenger produced by PLD, reverses ETOH inhibition of NSC proliferation. Blocking the PLD signal by 1-butanol suppresses the proliferation. ETOH-induced suppression of NSC proliferation and the protective effect of PA for ETOH-induced suppression are mediated through extracellular signal-regulated kinase signaling. These results indicate that exposure to ETOH impairs NSC proliferation by altering the PLD signaling pathway.


Subject(s)
Embryonic Stem Cells/drug effects , Ethanol/toxicity , Neurons/drug effects , Phospholipase D/antagonists & inhibitors , 1-Butanol/pharmacology , Animals , Apoptosis , Cell Proliferation/drug effects , Cells, Cultured , Embryonic Stem Cells/enzymology , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Ethanol/antagonists & inhibitors , Gene Expression/drug effects , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Neurons/enzymology , Phosphatidic Acids/antagonists & inhibitors , Phosphatidic Acids/metabolism , Phosphatidic Acids/pharmacology , Phospholipase D/genetics , Phospholipase D/metabolism , Up-Regulation
8.
Steroids ; 73(2): 216-21, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18036628

ABSTRACT

In the present study the effects of diethylstilbestrol on phospholipase D activity and degranulation by human neutrophils were examined. Diethylstilbestrol is a synthetic estrogen and has structural similarity to resveratrol. Resveratrol is a natural polyphenolic antioxidant and has been shown to inhibit the activity of phospholipase D in stimulated neutrophils. Phospholipase D catalyzes the hydrolysis of phosphatidylcholine to yield phosphatidic acid and choline. It also catalyzes the transfer of the phosphatidyl group to ethanol forming phosphatidylethanol at the expense of phosphatidic acid. Phospholipase D activation is associated with degranulation by neutrophils stimulated with chemotactic peptide, formyl-methionyl-leucyl-phenylalanine. The results show that diethylstilbestrol at 100 microM induced a complete inhibition of phosphatidic acid formation in neutrophils, the latter activated by chemotactic peptide. In the presence of ethanol, diethylstilbestrol dose dependently reduced phosphatidylethanol formation induced by chemotactic peptide or by phorbol 12-myristate 13-acetate, indicative of diethylstilbestyrol inhibition of phospholipase D activity. The results also demonstrate that diethylstilbestrol inhibited degranulation by chemotactic peptide-stimulated neutrophils. In comparison to resveratrol, diethylstilbestrol exhibits a stronger inhibition on PA formation, phospholipase D activity and degranulation. These findings suggest that diethylstilbestrol-like resveratrol, may have anti-inflammatory effect in vitro.


Subject(s)
Cell Degranulation/drug effects , Diethylstilbestrol/pharmacology , Phospholipase D/drug effects , Anti-Inflammatory Agents , Cells, Cultured , Dose-Response Relationship, Drug , Humans , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophil Activation , Neutrophils , Phosphatidic Acids/antagonists & inhibitors
9.
J Cell Sci ; 119(Pt 8): 1645-54, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16569667

ABSTRACT

Phospholipase D catalyzes the hydrolysis of phosphatidylcholine to generate phosphatidic acid, and there is currently much interest in elucidating messenger functions for this molecule. We report here that wounding sheets of corneal epithelial and Madin Darby canine kidney cells induces strong activation of phospholipase D, and we provide evidence that activation is amplified through a positive feed-back loop. Short-chain analogues of phosphatidic acid induce motility robustly in corneal and other epithelial cell types. The effects of these analogues were not the result of their conversion to the corresponding diacylglycerol or lysophosphatidic acid, implying that phosphatidic acid acts directly on one or more cellular targets. Strikingly, phosphatidic acid signaling was found to stimulate the epidermal growth factor receptor (EGFR) through a transactivation process. Healing of wounds in sheets of corneal epithelial cells is absolutely dependent on epidermal growth factor receptor signaling, and the present data suggest that its activation is a result of wound-induced phospholipase D activation.


Subject(s)
Cell Movement , Epithelium, Corneal/metabolism , ErbB Receptors/metabolism , Phosphatidic Acids/pharmacology , Phospholipase D/metabolism , Animals , Cell Adhesion Molecules/metabolism , Cells, Cultured , Dogs , Dose-Response Relationship, Drug , Enzyme Activation , Epidermal Growth Factor/pharmacology , Humans , In Vitro Techniques , Models, Biological , Phosphatidic Acids/antagonists & inhibitors , Protein Kinase C/metabolism , Rabbits , Signal Transduction , Wound Healing
10.
J Immunol ; 174(9): 5201-8, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15843515

ABSTRACT

Activation of phospholipase D (PLD) and protein kinase C (PKC) as well as calcium mobilization are essential signals for degranulation of mast cells. However, the exact role of PLD in degranulation remains undefined. In this study we have tested the hypothesis that the PLD product, phosphatidic acid, and diacylglycerides generated therefrom might promote activation of PKC. Studies were conducted in two rodent mast cell lines that were stimulated with Ag via FcepsilonRI and a pharmacologic agent, thapsigargin. Diversion of production of phosphatidic acid to phosphatidylbutanol (the transphosphatidylation reaction) by addition of l-butanol suppressed both the translocation of diacylglyceride-dependent isoforms of PKC to the membrane and degranulation. Tertiary-butanol, which is not a substrate for the transphosphatidylation, had a minimal effect on PKC translocation and degranulation, and 1-butanol itself had no effect on PKC translocation when PKC was stimulated directly with phorbol ester, 12-O-tetradecanoylphorbol-13-acetate. Also, in cells transfected with small inhibitory RNAs directed against PLD1 and PLD2, activation of PLD, generation of diacylglycerides, translocation of PKC, and degranulation were all suppressed. Phorbol ester, which did not stimulate degranulation by itself, restored degranulation when used in combination with thapsigargin whether PLD function was disrupted with 1-butanol or the small inhibitory RNAs. However, degranulation was not restored when cells were costimulated with Ag and phorbol ester. These results suggested that the production of phosphatidic acid by PLD facilitates activation of PKC and, in turn, degranulation, although additional PLD-dependent processes appear to be critical for Ag-mediated degranulation.


Subject(s)
Cell Degranulation/immunology , Mast Cells/enzymology , Phospholipase D/physiology , Protein Kinase C/metabolism , 1-Butanol/antagonists & inhibitors , 1-Butanol/pharmacology , Animals , Cell Degranulation/drug effects , Cell Line, Transformed , Cell Line, Tumor , Diglycerides/antagonists & inhibitors , Diglycerides/biosynthesis , Drug Synergism , Enzyme Activation/drug effects , Enzyme Activation/immunology , Enzyme Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Phosphatidic Acids/antagonists & inhibitors , Phosphatidic Acids/biosynthesis , Phospholipase D/antagonists & inhibitors , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Transport/drug effects , Protein Transport/immunology , RNA, Small Interfering/pharmacology , Rats , Tetradecanoylphorbol Acetate/pharmacology , Thapsigargin/pharmacology
11.
J Cell Biochem ; 92(3): 481-90, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15156560

ABSTRACT

The secretion of matrix metalloproteinase (MMP-9) is stimulated by the glucocorticoid-induced tumor necrosis factor receptor (GITR), a new tumor necrosis factor receptor (TNFR) family, in murine macrophages via an activation of protein kinase C (PKC)delta and phospholipase D (PLD). Secretions of MMP-9 are stimulated by the phosphatidic acid (PA), a product of PLD activity and an inhibition of PA production by a 1-propanol inhibited secretion of MMP-9 by soluble GITR (sGITR). MMP-9 is not secreted by diacylglycerol (DAG) and an inhibitor of PA phosphatase has no effect on the secretion induced by sGITR, indicating that PA is responsible for MMP-9 secretion in murine macrophages. Our data indicates that sGITR-induced activation of PKCdelta and PLD increases MMP-9 secretions in macrophages.


Subject(s)
Macrophages/enzymology , Macrophages/metabolism , Matrix Metalloproteinase 9/metabolism , Phospholipase D/metabolism , Protein Kinase C/metabolism , Receptors, Nerve Growth Factor/metabolism , Receptors, Tumor Necrosis Factor/metabolism , 1-Propanol/pharmacology , Animals , Cell Line , Diglycerides/pharmacology , Enzyme Activation , Glucocorticoid-Induced TNFR-Related Protein , Mice , Phosphatidate Phosphatase/antagonists & inhibitors , Phosphatidate Phosphatase/metabolism , Phosphatidic Acids/antagonists & inhibitors , Phosphatidic Acids/pharmacology , Protein Kinase C-delta , Solubility
12.
J Neurochem ; 88(3): 680-8, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14720217

ABSTRACT

Receptor phosphorylation, arrestin binding, uncoupling from G protein and subsequent endocytosis have been implicated in G protein-coupled receptor desensitization after chronic agonist exposure. In search of proteins regulating the mu-opioid receptor endocytosis, we have recently established that activation of phospholipase D (PLD)2 is required for agonist-induced mu-opioid receptor endocytosis. In this study, we determined the effect of PLD2 activity on the desensitization and resensitization rate of the mu-opioid receptor. We clearly demonstrated that inhibition of PLD2-mediated phosphatidic acid formation by alcohol (1-butanol or ethanol) or overexpression of a dominant negative mutant of PLD2 prevented agonist-mediated endocytosis and resulted in a faster desensitization rate of the mu-opioid receptor after chronic (D-Ala2, Me Phe4, Glyol5)enkephalin treatment in human embryonic kidney 293 cells. Moreover, inhibition of PLD2 activity led to an impairment of the resensitization rate of the mu-opioid receptor. In summary, our data strongly suggest that PLD2 is a modulator of agonist-induced endocytosis, desensitization and resensitization of the mu-opioid receptor.


Subject(s)
Phospholipase D/physiology , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , 1-Butanol/pharmacology , Analgesics, Opioid/pharmacology , Animals , Cell Line , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Humans , Phosphatidic Acids/antagonists & inhibitors , Phosphatidic Acids/biosynthesis , Phospholipase D/antagonists & inhibitors , Protein Isoforms/agonists , Protein Isoforms/metabolism , Rats
14.
Arterioscler Thromb Vasc Biol ; 20(12): E127-33, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11116077

ABSTRACT

In the present study, the roles of the small GTPase RhoA and its target Rho kinase in endothelial permeability were investigated in vitro. We have shown previously that, in addition to a rise in the intracellular Ca(2+) concentration ([Ca(2+)](i)), RhoA is involved in the prolonged thrombin-induced barrier dysfunction. To study the role of RhoA and Rho kinase more specifically, endothelial cells were stimulated with lysophosphatidic acid (LPA), a commonly used RhoA activator. LPA induced a 2- to 3-fold increase in the passage of horseradish peroxidase (HRP) across endothelial monolayers that lasted for several hours, whereas thrombin induced a 5- to 10-fold increase. Comparable to the thrombin-induced barrier dysfunction, the LPA-induced barrier dysfunction was accompanied by a reorganization of the F-actin cytoskeleton and the formation of focal attachment sites. LPA induced only a transient increase in myosin light-chain (MLC) phosphorylation, which returned to basal level within 10 minutes. In endothelial cells, [Ca(2+)](i) was not elevated by LPA. Chelation of Ca(2+)(i) ions by 1, 2-bis(2-aminophenoxy)ethane-N:,N:,N:',N:'-tetraacetic acid did not prevent the LPA-induced passage of HRP. Apparently, a low degree of MLC kinase activation occurred, because the MLC kinase inhibitor KT5926 reduced the levels of both basal and LPA-stimulated HRP passage. Inhibition of RhoA by the C3 transferase from Clostridium botulinum inhibited the LPA-induced cytoskeletal changes and prevented the LPA-induced HRP passage. Inhibition of Rho kinase by Y-27632 completely prevented the LPA-induced increase in HRP passage without affecting basal permeability. These data indicate that LPA-induced endothelial hyperpermeability occurs without a change in [Ca(2+)](i) and requires activation of RhoA and Rho kinase.


Subject(s)
Endothelium, Vascular/metabolism , Protein Serine-Threonine Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Amides/pharmacology , Calcium/metabolism , Cell Membrane Permeability/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Cytoskeleton/drug effects , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Horseradish Peroxidase , Humans , Intracellular Signaling Peptides and Proteins , Myosin Light Chains/metabolism , Phosphatidic Acids/antagonists & inhibitors , Phosphatidic Acids/pharmacology , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/pharmacology , Thrombin/pharmacology , Time Factors , Umbilical Veins , rho-Associated Kinases , rhoA GTP-Binding Protein/antagonists & inhibitors
15.
Dent Mater ; 16(4): 297-302, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10831786

ABSTRACT

OBJECTIVES: Diacylglycerol-kinase (DAG-kinase) is an enzyme that phosphorylates diacylglycerol (DAG) to phosphatidic acid (PA), which serves as a precursor to phosphoglycerides involved in cell signaling or as cell membrane structural components. DAG-kinase can be inhibited by diacylethylene glycols (DAEG). We hypothesize that 2-hydroxyethyl methacrylate (HEMA) may alter phosphorylation of DAG to PA following intracellular formation of DAEG. METHODS: Cultured rabbit kidney (RK13) epithelial cells were treated with HEMA, EG, or known inhibitors of DAG-kinase for 24 h, then exposed to [32P]O4- in the presence of a synthetic diacylglycerol for 2 h. Other cultures were radiolabeled with [3H]-oleic acid for 24 h, then exposed to HEMA for an additional 24 h. The cells were harvested and the lipids extracted. Radioactive lipids were separated by thin layer chromatography, located by autoradiography, and quantitated as cpm/ug protein. Cell cultures treated with HEMA were homogenized and the DAG-kinase activity was assayed and expressed as cpm/ug protein. Data were analyzed by one-way ANOVA and Newman-Keuls Multiple Comparison Test. RESULTS: Cultures exposed to HEMA or known DAG-kinase inhibitors exhibited reduced incorporation of radioactivity in the PA fraction compared to control cultures. Direct assays of DAG-kinase activity from cells exposed to HEMA demonstrated decreased enzyme activity. Evaluation of cell phospholipid synthesis showed altered formation of phosphatidylethanolamine and phosphatidylcholine. SIGNIFICANCE: Results suggest that HEMA impairs formation of PA, possibly by acylation of EG released by hydrolysis of the HEMA and resultant production of the inhibitor DAEG. The decreased availability of PA may alter PA-dependent cell structural lipid pathways and lipid-dependent signaling pathways, altering cell growth.


Subject(s)
Dentin-Bonding Agents/pharmacology , Methacrylates/pharmacology , Phosphatidic Acids/metabolism , Analysis of Variance , Animals , Autoradiography , Cells, Cultured , Chromatography, Thin Layer , Diacylglycerol Kinase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Hydrolysis , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Oleic Acid/metabolism , Phosphatidic Acids/antagonists & inhibitors , Phosphatidylcholines/metabolism , Phosphatidylethanolamines/metabolism , Phosphorylation , Rabbits , Radiopharmaceuticals , Statistics as Topic , Tritium
16.
Transplantation ; 63(12): 1840-3, 1997 Jun 27.
Article in English | MEDLINE | ID: mdl-9210515

ABSTRACT

We have previously shown the existence of migratory hematopoietic stem cells in adult solid organs. This study demonstrates that granulocyte colony-stimulating factor (G-CSF) and lisofylline, a phosphatidic acid inhibitor that suppresses hematopoiesis-inhibiting cytokines, can enhance the engraftment of organ-based hematopoietic stem cells. When syngeneic heart grafts or liver nonparenchymal cells were transplanted into lethally irradiated (9.5 Gy) Lewis rats, complete hematopoietic reconstitution and animal survival were significantly improved by treating the recipient with G-CSF or, to a lesser extent, with lisofylline. Pretreatment of hepatic nonparenchymal cell donors with G-CSF, but not lisofylline, also resulted in striking improvement of recipient survival which was associated with an augmented subpopulation of donor stem cells. The results suggest that these drugs can be used to enhance the chimerism that we postulate to be the basis of organ allograft acceptance.


Subject(s)
Adjuvants, Immunologic/pharmacology , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Leukocytes/drug effects , Pentoxifylline/analogs & derivatives , Phosphatidic Acids/antagonists & inhibitors , Whole-Body Irradiation , Animals , Bone Marrow Transplantation , Heart Transplantation , Liver Transplantation , Pentoxifylline/pharmacology , Rats , Rats, Inbred Lew
17.
Am J Respir Crit Care Med ; 155(3): 928-36, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9117028

ABSTRACT

The effects of lisofylline [(R)-1-(5-hydroxyhexyl)-3,7-dimethylxanthine] (LSF), an inhibitor of de novo phosphatidic acid (PA) generation, on sepsis-induced acute lung injury was studied using Hanford minipigs weighing 18 to 25 kg. Sepsis was induced by an intravenous infusion of Pseudomonas aeruginosa (1 x 10(6)/colony-forming units/kg/min over 2 h). Saline was used as the control vehicle. Six groups were studied: saline control group (SALINE: n = 5); sepsis control group (SEPSIS: n = 5); LSF control group (LSF: n = 5), which received a 25-mg/kgbolus of LSF 30 min before time zero followed by continuous infusion of 10 mg/kg/h throughout the study; LSF-treated septic groups, which were treated with LSF 30 min prior to sepsis (Pre: n = 5), 1 h postonset (Post-1 h: n = 8) or h postonset (Post-2 h: n = 8) of the bacterial infusion. Hemodynamics PaO2, neutrophil counts, and plasma porcine tumor necrosis factor-alpha concentrations were monitored for 6 h. After the minipigs were killed, lung tissue was sampled to measured wet-to-dry weight ratio (W/D), tissue albumin index (TAI), thiobarbituric acid-reactive material content (TBARM), and myeloperoxidase (MPO) activity. Compared with the SALINE group, the SEPSIS group showed significant systemic hypotension, pulmonary hypertension, arterial hypoxemia, neutropenia, and increase in TNF-alpha, MPO activity, W/D, TBARM, and TAI. LSF treatment attenuated sepsis-induced pulmonary hypertension, neutropenia, and hypoxemia, and increased MPO activity and lung injury measurements in the Pre and Post-1 h groups, but its efficacy was blunted in the Post-2 h group. Plasma TNF-alpha was decreased only in the Pre group. Thus, inhibition of intracellular PA generation through de novo pathways attenuates sepsis-induced acute lung injury.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Pentoxifylline/analogs & derivatives , Phosphatidic Acids/antagonists & inhibitors , Respiratory Distress Syndrome/prevention & control , Shock, Septic/prevention & control , Adjuvants, Immunologic/administration & dosage , Animals , Disease Models, Animal , Female , Hemodynamics , Lung/pathology , Male , Pentoxifylline/administration & dosage , Pentoxifylline/therapeutic use , Pseudomonas Infections , Respiratory Distress Syndrome/etiology , Shock, Septic/microbiology , Shock, Septic/physiopathology , Swine , Swine, Miniature , Time Factors
18.
Endocrinology ; 137(12): 5421-8, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8940366

ABSTRACT

The present study was undertaken to determine whether phospholipase D participates in the mitogenic action of arginine vasopressin (AVP) in cultured rat glomerular mesangial cells. AVP promptly increased the phosphatidylethanol formation in a concentration-dependent manner, which indicates the activation of phospholipase D. When cells were preincubated with 2,3-diphosphoglycerate or carbobenzyloxy-leucine-tyrosine-chloromethylketone (zLYCK), inhibitors of phospholipase D, the 1 x 10(-7) M AVP-produced phosphatidylethanol was significantly attenuated. Also, inhibitors of protein kinase C, staurosporine and calphostin C, reduced the AVP-induced increase in phosphatidylethanol. AVP activated mitogen-activated protein (MAP) kinase in a concentration-dependent manner. Such an activation was significantly reduced by 2,3-diphosphoglycerate, zLYCK, or staurosporine. Also, AVP stimulated [3H]thymidine incorporation, an effect significantly less in the presence of 2,3-diphosphoglycerate or zLYCK. Similar results were obtained with exogenous bacterial phospholipase D. Both MAP kinase and [3H]thymidine incorporation were not altered by 2,3-diphosphoglycerate or zLYCK per se. These results indicate that AVP activates phospholipase D and promotes cellular growth mediated through phospholipase D, in addition to a phospholipase C-dependent signal transduction, in glomerular mesangial cells.


Subject(s)
Arginine Vasopressin/pharmacology , Glomerular Mesangium/drug effects , Glomerular Mesangium/metabolism , Glycerophospholipids , Mitogens/pharmacology , Phospholipase D/metabolism , 2,3-Diphosphoglycerate , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Diphosphoglyceric Acids/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Glomerular Mesangium/cytology , Male , Naphthalenes/pharmacology , Phosphatidic Acids/antagonists & inhibitors , Phosphatidic Acids/metabolism , Phospholipase D/antagonists & inhibitors , Phospholipase D/pharmacology , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Staurosporine/pharmacology , Tetradecanoylphorbol Acetate/pharmacology
19.
Endocrinology ; 137(11): 4871-7, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8895359

ABSTRACT

Interleukin-1 beta (IL-1 beta) causes rat islet cell dysfunction through mechanisms that involve inducible nitric oxide synthase (iNOS). However, IL-1 beta also activates several lipid pathways, including those generating phosphatidic acid (PA). Lisofylline (LSF), a water-soluble, nontoxic, selective inhibitor of the PA-1 alpha subspecies, which is stimulated by IL-1 beta and tumor necrosis factor-alpha, has been shown to prevent cytokine-induced cytotoxicity in in vivo animal models. To evaluate the effect of LSF on acute IL-1 beta-induced islet dysfunction, rat islets were exposed to IL-1 beta (0.1 ng/ml) with or without LSF (100 microM) for 24 h, followed by 25 mM glucose (G) stimulation, measurement of rat insulin by RIA, and calculation of the insulin secretion rate. In other experiments, rat islets were precultured for 48 h, then treated for 48 h in 25 mM G with or without IL-1 beta (0.1 ng/ml) and LSF (400 microM), and aliquots of medium were removed at 0, 24, and 48 h for measurement of rat insulin. In addition, islets were exposed to 25 mM G with or without IL-1 beta and LSF, lipids were then extracted, and PA subspecies were identified by TLC and mass spectroscopy, and quantitated using normal phase HPLC. Islets were also exposed to IL-1 beta with or without LSF, and Western immunoblots were performed to evaluate the effect of LSF on iNOS protein expression. IL-1 beta caused a 44% decrease in islet G-stimulated insulin secretion compared to that in untreated islets (P < 0.0005), which was totally reversed by LSF. In addition, IL-1 beta decreased the G-stimulated medium insulin content by 75% at 24 h (P = 0.0004) and 86% at 48 h compared to that in control islets (P < 0.0001). LSF-treated islets maintained 70% of medium insulin content at 24 h (P = 0.11) and 50% at 48 h (P < 0.0001) compared to control islets. HPLC quantitation of PA-1 alpha extracted from islets treated with IL-1 beta alone showed an approximately 15-fold increase over the PA-1 alpha content of islets treated with IL-1 beta and LSF. IL-1 beta-induced expression of iNOS was unchanged with the addition of LSF. These results suggest that LSF is effective in reducing IL-1 beta-induced islet dysfunction, thus supporting the role of lipid mediators such as PA in cytokine-induced islet toxicity.


Subject(s)
Insulin/metabolism , Interleukin-1/pharmacology , Islets of Langerhans/physiology , Pentoxifylline/analogs & derivatives , Phosphatidic Acids/biosynthesis , Adjuvants, Immunologic/pharmacology , Analysis of Variance , Animals , Cells, Cultured , Chromatography, High Pressure Liquid , Drug Interactions , Glucose/pharmacology , Insulin Secretion , Interleukin-1/antagonists & inhibitors , Islets of Langerhans/drug effects , Male , Pentoxifylline/pharmacology , Phosphatidic Acids/antagonists & inhibitors , Phosphatidic Acids/chemistry , Protein Biosynthesis , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Fast Atom Bombardment
SELECTION OF CITATIONS
SEARCH DETAIL
...