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










Publication year range
1.
J Immunol ; 180(10): 6977-87, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18453620

ABSTRACT

IL-1 is a major proinflammatory cytokine which interacts with the IL-1 receptor I (IL-1RI) complex, composed of IL-1RI and IL-1R accessory protein subunits. Currently available strategies to counter pathological IL-1 signaling rely on a recombinant IL-1 receptor antagonist, which directly competes with IL-1 for its binding site. Presently, there are no small antagonists of the IL-1RI complex. Given this void, we derived 15 peptides from loops of IL-1R accessory protein, which are putative interactive sites with the IL-1RI subunit. In this study, we substantiate the merits of one of these peptides, rytvela (we termed "101.10"), as an inhibitor of IL-1R and describe its properties consistent with those of an allosteric negative modulator. 101.10 (IC(50) approximately 1 nM) blocked human thymocyte proliferation in vitro, and demonstrated robust in vivo effects in models of hyperthermia and inflammatory bowel disease as well as topically in contact dermatitis, superior to corticosteroids and IL-1ra; 101.10 did not bind to IL-1RI deficient cells and was ineffective in vivo in IL-1RI knockout mice. Importantly, characterization of 101.10, revealed noncompetitive antagonist actions and functional selectivity by blocking certain IL-1R pathways while not affecting others. Findings describe the discovery of a potent and specific small (peptide) antagonist of IL-1RI, with properties in line with an allosteric negative modulator.


Subject(s)
Anti-Inflammatory Agents , Inflammation/drug therapy , Interleukin 1 Receptor Antagonist Protein , Peptides/chemistry , Peptides/pharmacology , Receptors, Interleukin-1/antagonists & inhibitors , Amino Acid Sequence , Animals , Blotting, Western , Dermatitis/drug therapy , Humans , Inflammatory Bowel Diseases/drug therapy , Mice , Mice, Knockout , Molecular Sequence Data , Peptides/chemical synthesis , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley
2.
Am J Physiol Regul Integr Comp Physiol ; 292(3): R1174-83, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17122328

ABSTRACT

Oxidant stress plays a significant role in hypoxic-ischemic injury to the susceptible microvascular endothelial cells. During oxidant stress, lysophosphatidic acid (LPA) concentrations increase. We explored whether LPA caused cytotoxicity to neuromicrovascular cells and the potential mechanisms thereof. LPA caused a dose-dependent death of porcine cerebral microvascular as well as human umbilical vein endothelial cells; cell death appeared oncotic rather than apoptotic. LPA-induced cell death was mediated via LPA(1) receptor, because the specific LPA(1) receptor antagonist THG1603 fully abrogated LPA's effects. LPA decreased intracellular GSH levels and induced a p38 MAPK/JNK-dependent inducible nitric oxide synthase (NOS) expression. Pretreatment with the antioxidant GSH precursor N-acetyl-cysteine (NAC), as well as with inhibitors of NOS [N(omega)-nitro-l-arginine (l-NNA); 1400W], significantly prevented LPA-induced endothelial cell death (in vitro) to comparable extents; as expected, p38 MAPK (SB203580) and JNK (SP-600125) inhibitors also diminished cell death. LPA did not increase indexes of oxidation (isoprostanes, hydroperoxides, and protein nitration) but did augment protein nitrosylation. Endothelial cytotoxicity by LPA in vitro was reproduced ex vivo in brain and in vivo in retina; THG1603, NAC, l-NNA, and combined SB-203580 and SP600125 prevented the microvascular rarefaction. Data implicate novel properties for LPA as a modulator of the cell redox environment, which partakes in endothelial cell death and ensued neuromicrovascular rarefaction.


Subject(s)
Astrocytes/drug effects , Endothelial Cells/drug effects , Lysophospholipids/pharmacology , Oxidation-Reduction/drug effects , Animals , Animals, Newborn , Astrocytes/cytology , Brain/blood supply , Brain/cytology , Cell Death , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Endothelial Cells/ultrastructure , Endothelium, Vascular/cytology , Kinetics , Models, Biological , Rats , Rats, Sprague-Dawley , Sus scrofa
3.
J Biol Chem ; 281(23): 16058-67, 2006 Jun 09.
Article in English | MEDLINE | ID: mdl-16574649

ABSTRACT

Stimulation of freshly isolated rat hepatocytes with lysophosphatidic acid (LPA) resulted in LPA1 receptor-mediated and nitricoxide-dependent up-regulation of the immediate early genes iNOS (inducible nitric-oxide synthase (NOS)) and mPGES-1 (microsomal prostaglandin E synthase-1). Because LPA is a ligand for both cell surface and intracellular receptor sites and a potent endothelial NOS (eNOS) activator, we hypothesized that NO derived from activated nuclearized eNOS might participate in gene regulation. Herein we show, by confocal microscopy performed on porcine cerebral endothelial cells expressing native LPA1-receptor and eNOS and on HTC4 rat hepatoma cells co-transfected with recombinant human LPA1-receptor and fused eNOS-GFP cDNA, a dynamic eNOS translocation from peripheral to nuclear regions upon stimulation with LPA. Nuclear localization of eNOS and its downstream effector, soluble guanylate cyclase, were demonstrated in situ in rat liver specimens by immunogold labeling using specific antibodies. Stimulation of this nuclear fraction with LPA and the NO donor sodium nitroprusside resulted, respectively, in increased production of nitrite (and eNOS phosphorylation) and cGMP; these separate responses were also correspondingly blocked by NOS inhibitor L-NAME and soluble guanylate cyclase inhibitor ODQ. In addition, sodium nitroprusside evoked a sequential increase in nuclear Ca2+ transients, activation of p42 MAPK, NF-kappaB binding to DNA consensus sequence, and dependent iNOS RNA. This study describes a hitherto unrecognized molecular mechanism by which nuclear eNOS through ensuing NO modulates nuclear calcium homeostasis involved in gene transcription-associated events. Moreover, our findings strongly support the concept of the nucleus as an autonomous signaling compartment.


Subject(s)
Genes, Immediate-Early , Inflammation/genetics , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide/metabolism , Prostaglandin-Endoperoxide Synthases/genetics , Signal Transduction , Animals , Base Sequence , Cell Nucleus/metabolism , Cells, Cultured , DNA Primers , Gene Expression/drug effects , Guanylate Cyclase , Humans , Liver/metabolism , Lysophospholipids/pharmacology , Microscopy, Confocal , Nitric Oxide Synthase Type III/genetics , Prostaglandin-E Synthases , Rats , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Lysophosphatidic Acid/genetics , Reverse Transcriptase Polymerase Chain Reaction , Soluble Guanylyl Cyclase , Swine
4.
Free Radic Biol Med ; 40(3): 543-53, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16443170

ABSTRACT

Neovascularization after an ischemic insult is a beneficial attempt to salvage the injured tissue. Yet, despite the production of angiogenic factors within ischemic tissues, compensatory growth of new vessels fails to provide adequate vascularization. Thus, we hypothesized that local factors counter efficient revascularization. Whereas ischemia is often considered to be synonymous with an oxygen deficit, it is also associated with a concomitant local elevation of carbon dioxide (CO2). Although studies suggest that hypercapnia impacts tissue neovascularization, its significance relative to the abundantly described effects of hypoxia and its underlying mechanisms have yet to be elucidated. Therefore, we investigated the effects of hypercapnia on blood vessel growth in models of developmental and ischemic neovascularization. Acute and prolonged CO2 exposure inhibited developmental neovascularization of the rodent retina, as well as revascularization of the ischemic retina. Hypercapnia induced early increases in endothelial nitric oxide synthase and nitrative stress, associated with astrocyte impairment and endothelial cell death, as well as downregulation of the proangiogenic prostaglandin E2 receptor EP3. These results establish a previously unexplored means by which hypercapnia hinders efficient neovascularization, a mechanism that may contribute to ischemic tissue injury.


Subject(s)
Hypercapnia , Neovascularization, Pathologic/prevention & control , Nitrates/metabolism , Oxidative Stress , Reactive Nitrogen Species/metabolism , Animals , Astrocytes/metabolism , Cell Death , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Ischemia/metabolism , Ischemia/pathology , Nitric Oxide Synthase Type III/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP3 Subtype , Retina/growth & development , Retinal Neovascularization/metabolism , Retinal Vessels/metabolism
5.
J Pharmacol Exp Ther ; 316(2): 618-27, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16214879

ABSTRACT

Thromboxane A(2) (TXA(2)) is an important lipid mediator generated during oxidative stress and implicated in ischemic neural injury. This autacoid was recently shown to partake in this injury process by directly inducing endothelial cytotoxicity. We explored the mechanisms for this TXA(2)-evoked neural microvascular endothelial cell death. Stable TXA(2) mimetics 5-heptenoic acid, 7-[6-(3-hydroxy-1-octenyl)-2-oxabicyclo[2.2.1]hept-5-yl]-[1R-[1alpha,4alpha,5beta(Z),6alpha,(1E,3S)]]-9,11-dedioxy-9alpha,11alpha-methanolpoxy (U-46619) [as well as [1S-[1alpha,2alpha(Z),3beta(1E,3S(*)),4alpha]]-7-[3-[3-hydroxy-4-(4-iodophenoxy)-1-butenyl]-7-oxabicyclo[2.1.1]-hept-2-yl]-5-heptenoic acid; I-BOP] induced a retinal microvascular degeneration in rat pups in vivo and in porcine retinal explants ex vivo and death of porcine brain endothelial cells (in culture). TXA(2) dependence of these effects was corroborated by antagonism using the selective TXA(2) receptor blocker (-)-6,8-difluoro-9-p-methyl-sulfonyl-benzyl-1,2,3,4-tetrahydrocarbazol-1-yl-acetic acid (L670596). In all cases, neurovascular endothelial cell death was prevented by pan-calpain and specific m-calpain inhibitors but not by caspase-3 or pan-caspase inhibitors. Correspondingly, TXA(2) (mimetics) augmented generation of known active m-calpain (but not mu-calpain) form and increased the activity of m-calpain (cleavage of fluorogenic substrate N-succinyl-Leu-Leu-Val-Tyr-7-amino-4-methylcoumarin; and of alpha-spectrin into specific fragments) but not of pan-caspase or specific caspase-3 (respectively, using sulforhodamine-Val-Arg-Asp-fluoromethyl ketone and detecting its active 17- and 12-kDa fragments). Interestingly, these effects were phospholipase C (PLC)-dependent [associated with increase in inositol triphosphate and inhibited by PLC blocker 1-[6-[[17beta-methoxyestra-1,3,5(10)-trien-17-yl]amino]hexyl]-1H-pyrrole-2,5-dione (U73122)] and required calcium but were not associated with increased intracellular calcium. U-46619-induced calpain activation resulted in translocation of Bax to the mitochondria, loss of polarization of the latter (using potentiometric probe 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolyl-carbocyanine iodide; JC-1) and in turn release of cytochrome c into the cytosol and depletion of cellular ATP; these effects were all blocked by calpain inhibitors. Overall, this work identifies (specifically) m-calpain as a dominant protease in TXA(2)-induced neurovascular endothelial cell death.


Subject(s)
Calpain/physiology , Endothelium, Vascular/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/antagonists & inhibitors , Retinal Degeneration/metabolism , Retinal Vessels/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Animals, Newborn , Calcium/metabolism , Calpain/metabolism , Carbazoles/pharmacology , Cell Survival/drug effects , Cytosol/drug effects , Cytosol/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Inositol Phosphates/metabolism , Membrane Potentials/drug effects , Microcirculation/drug effects , Microcirculation/metabolism , Microcirculation/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Rats , Rats, Sprague-Dawley , Retinal Degeneration/chemically induced , Retinal Degeneration/pathology , Retinal Vessels/drug effects , Retinal Vessels/pathology
6.
Nat Med ; 11(12): 1339-45, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16311602

ABSTRACT

Nitrative stress has an important role in microvascular degeneration leading to ischemia in conditions such as diabetic retinopathy and retinopathy of prematurity. Thus far, mediators of nitrative stress have been poorly characterized. We recently described that trans-arachidonic acids are major products of NO(2)(*)-mediated isomerization of arachidonic acid within the cell membrane, but their biological relevance is unknown. Here we show that trans-arachidonic acids are generated in a model of retinal microangiopathy in vivo in a NO(*)-dependent manner. They induce a selective time- and concentration-dependent apoptosis of microvascular endothelial cells in vitro, and result in retinal microvascular degeneration ex vivo and in vivo. These effects are mediated by an upregulation of the antiangiogenic factor thrombospondin-1, independently of classical arachidonic acid metabolism. Our findings provide new insight into the molecular mechanisms of nitrative stress in microvascular injury and suggest new therapeutic avenues in the management of disorders involving nitrative stress, such as ischemic retinopathies and encephalopathies.


Subject(s)
Apoptosis/physiology , Arachidonic Acids/toxicity , Diabetic Angiopathies/metabolism , Endothelial Cells/drug effects , Gene Expression Regulation/drug effects , Neovascularization, Physiologic/drug effects , Retinal Vessels/cytology , Thrombospondin 1/metabolism , Animals , Apoptosis/drug effects , Arachidonic Acids/metabolism , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , DNA Primers , In Situ Nick-End Labeling , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley , Retinal Vessels/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Sus scrofa , Tetrazolium Salts , Thiazoles
7.
Free Radic Biol Med ; 37(3): 358-66, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15223069

ABSTRACT

Oxidant stress plays a significant role in the pathogenesis of periventricular leukomalacia (PVL). Isoprostanes (IsoPs) are bioactive products of lipid peroxidation abundantly generated during hypoxic-ischemic injuries. Because loss of oligodendrocytes (OLs) occurs early in PVL, we hypothesized that IsoPs could induce progenitor OL death. 15-E(2t)-IsoP but not 15-F(2t)-IsoP elicited a concentration-dependent death of progenitor OLs by oncosis and not by apoptosis, but exerted minimal effects on mature OLs. 15-E(2t)-IsoP-induced cytotoxicity could not be explained by its conversion into cyclopentenones, because PGA(2) was hardly cytotoxic. On the other hand, thromboxane A(2) (TxA(2)) synthase inhibitor CGS12970 and cyclooxygenase inhibitor ibuprofen attenuated 15-E(2t)-IsoP-induced cytotoxicity. Susceptibility of progenitor OLs was independent of TxA(2) receptor (TP) expression, which was far less in progenitor than in mature OLs. However, TxA(2) synthase was detected in precursor but not in mature OLs, and TxA(2) mimetic U46619 induced hydroperoxides generation and progenitor OL death. The glutathione synthesis enhancer N-acetylcysteine prevented 15-E(2t)-IsoP-induced progenitor cell death. Depletion of glutathione in mature OLs with buthionine sulfoximine rendered them susceptible to cytotoxicity of 15-E(2t)-IsoP. These novel data implicate 15-E(2t)-IsoP as a product of oxidative stress that may contribute in the genesis of PVL.


Subject(s)
Isoprostanes/toxicity , Oligodendroglia/cytology , Oligodendroglia/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Animals , Animals, Newborn , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Female , Male , Oligodendroglia/metabolism , Oxidative Stress , Prostaglandins A/metabolism , Rats , Rats, Sprague-Dawley , Thromboxane A2/metabolism
8.
Free Radic Biol Med ; 36(2): 163-72, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14744628

ABSTRACT

F2-isoprostanes (F2-IsoP's) are biologically active prostanoids formed by free radical-mediated peroxidation of arachidonic acid. Four different F2-IsoP regioisomers (5-, 8-, 12-, and 15-series), each comprising eight racemic diastereomers, total 64 compounds. Information regarding the biological activity of IsoP's is largely limited to 15-F2t-IsoP (8-iso-PGF2alpha). We recently demonstrated that 15-F2t-IsoP and its metabolite, 2,3-dinor-5,6-dihydro-15-F2t-IsoP, evoked vasoconstriction and TXA2 generation in retina and brain microvasculature. We have now examined and compared the biological activities of a series of recently synthesized new 5-, 12-, and 15-series F2-IsoP isomers in pig retinal and brain microvasculature. We hereby show that other 15-series F2-IsoP isomers, 15-epi-15-F2t-IsoP, ent-15-F2t-IsoP, and ent-15-epi-15-F2t-IsoP, are also potent vasoconstrictors. The 12-series isomers tested, 12-F2t-IsoP and 12-epi-12-F2t-IsoP, also caused marked vasoconstriction. Of the 5-series isomers tested, 5-F2t-IsoP and 5-epi-5-F2t-IsoP possessed no vasomotor properties, whereas ent-5-F2t-IsoP caused modest vasoconstriction. The vasoconstriction of ent-5-F2t-IsoP, 12-F2t-IsoP, and 12-epi-12-F2t-IsoP was abolished by removal of the endothelium, by TXA2 synthase and receptor inhibitor (CGS12970, L670,596), and by receptor-mediated Ca2+ channel blockade (SK & F96365); correspondingly, these isomers increased TXB2 formation by activating Ca2+ influx (detected with fura 2-AM) through non-voltage-dependent receptor-mediated Ca2+ entry (SK & F96365 sensitive) in endothelial cells. In conclusion, as seen with 15-F2t-IsoP, ent-5-F2t-IsoP, 12-F2t-IsoP, and 12-epi-12-F2t-IsoP constricted both retinal and brain microvessels by inducing endothelium-dependent TXA2 synthesis. These new findings broaden the scope of our understanding regarding the potential involvement of F2-IsoP's as mediators of oxidant injury.


Subject(s)
Brain/blood supply , F2-Isoprostanes/chemistry , F2-Isoprostanes/pharmacology , Microcirculation/drug effects , Retina/physiology , Vasoconstriction/drug effects , Animals , Calcium/metabolism , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , F2-Isoprostanes/chemical synthesis , In Vitro Techniques , Isomerism , Microcirculation/physiology , Swine , Thromboxane A2/metabolism
9.
J Biol Chem ; 278(40): 38875-83, 2003 Oct 03.
Article in English | MEDLINE | ID: mdl-12847111

ABSTRACT

Lysophosphatidic acid (LPA) is a bioactive molecule involved in inflammation, immunity, wound healing, and neoplasia. Its pleiotropic actions arise presumably by interaction with their cell surface G protein-coupled receptors. Herein, the presence of the specific nuclear lysophosphatidic acid receptor-1 (LPA1R) was revealed in unstimulated porcine cerebral microvascular endothelial cells (pCMVECs), LPA1R stably transfected HTC4 rat hepatoma cells, and rat liver tissue using complementary approaches, including radioligand binding experiments, electron- and cryomicroscopy, cell fractionation, and immunoblotting with three distinct antibodies. Coimmunoprecipitation studies in enriched plasmalemmal fractions of unstimulated pCMVEC showed that LPA1Rs are dually sequestrated in caveolin-1 and clathrin subcompartments, whereas in nuclear fractions LPA1R appeared primarily in caveolae. Immunofluorescent assays using a cell-free isolated nuclear system confirmed LPA1R and caveolin-1 co-localization. In pCMVEC, LPA-stimulated increases in cyclooxygenase-2 and inducible nitric-oxide synthase RNA and protein expression were insensitive to caveolea-disrupting agents but sensitive to LPA-generating phospholipase A2 enzyme and tyrosine kinase inhibitors. Moreover, LPA-induced increases in Ca2+ transients and/or iNOS expression in highly purified rat liver nuclei were prevented by pertussis toxin, phosphoinositide 3-kinase/Akt inhibitor wortmannin and Ca2+ chelator and channel blockers EGTA and SK&F96365, respectively. This study describes for the first time the nucleus as a potential organelle for LPA intracrine signaling in the regulation of pro-inflammatory gene expression.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/physiology , Receptors, G-Protein-Coupled , Androstadienes/pharmacology , Animals , Blotting, Western , Calcium/metabolism , Caveolin 1 , Caveolins/metabolism , Cell-Free System/metabolism , Cells, Cultured , Chelating Agents/pharmacology , Clathrin/metabolism , Egtazic Acid/pharmacology , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Immunoblotting , Liver/metabolism , Microcirculation , Microscopy, Electron , Microscopy, Fluorescence , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Pertussis Toxin/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phospholipases A/metabolism , Phospholipases A2 , Precipitin Tests , Protein Binding , Protein-Tyrosine Kinases/metabolism , Rats , Receptors, Lysophosphatidic Acid , Subcellular Fractions/metabolism , Swine , Time Factors , Transfection , Tumor Cells, Cultured , Wortmannin
10.
Stroke ; 34(3): 776-82, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12624307

ABSTRACT

BACKGROUND AND PURPOSE: Free radical-induced peroxidation is an important factor in the genesis of hypoxic-ischemic encephalopathy, including that of the preterm infant. Isoprostanes are major peroxidation products. Since microvascular dysfunction seems to contribute to ischemic encephalopathies, we studied the cytotoxicity of 8-iso-prostaglandin F2alpha (PGF2alpha) on cerebral microvascular cells. METHODS: Microvascular endothelial, astroglial, and smooth muscle cells from newborn brain were cultured. The cytotoxicity of 8-iso-PGF2alpha on these cells was determined by MTT assays and lactate dehydrogenase (LDH) release, propidium iodide incorporation, and DNA fragmentation (terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling [TUNEL]). In addition, effects of intraventricular injections of 8-iso-PGF2alpha and possible involvement of thromboxane in 8-iso-PGF2alpha-induced cytotoxicity were determined. RESULTS: 8-Iso-PGF2alpha induced time- and concentration-dependent endothelial cell death (EC50=0.1 nmol/L) but exerted little effect on smooth muscle and astroglial cells; endothelial cell death seemed mostly of oncotic nature (propidium iodide incorporation and LDH release). Cell death was associated with increased endothelial thromboxane A2 (TXA2) formation and was prevented by TXA2 synthase inhibitors (CGS12970 and U63557A); TXA2 mimetics U46619 and I-BOP also caused endothelial cell death. Intraventricular injection of 8-iso-PGF2alpha induced periventricular damage, which was attenuated by CGS12970 pretreatment. CONCLUSIONS: These data disclose a novel action of 8-iso-PGF2alpha involving TXA2 in oxidant stress-induced cerebral microvascular injury and brain damage.


Subject(s)
Brain Ischemia/metabolism , Brain/blood supply , Dinoprost/analogs & derivatives , Dinoprostone/analogs & derivatives , Endothelium, Vascular/drug effects , F2-Isoprostanes/pharmacology , Microcirculation/drug effects , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Brain/drug effects , Brain/pathology , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , DNA Fragmentation/drug effects , Dinoprostone/pharmacology , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Injections, Intraventricular , Isoprostanes/pharmacology , L-Lactate Dehydrogenase/metabolism , Microcirculation/cytology , Microcirculation/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Necrosis , Rats , Rats, Sprague-Dawley , Swine , Thromboxane A2/metabolism , Thromboxane-A Synthase/antagonists & inhibitors
11.
Am J Physiol Regul Integr Comp Physiol ; 284(4): R928-35, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12626359

ABSTRACT

Oxidant stress contributes to the pathogenesis of hypoxic-ischemic encephalopathies. Platelet-activating factor (PAF) is generated during oxidant stress. We studied the vasomotor mode of actions of PAF on periventricular (PV) microvessels of fetal ( approximately 75% of term), newborn (1-3 days), and adult pigs. PAF constricted PV microvessels from fetal (29.27 +/- 2.6%) and newborn (22.14 +/- 3.2%) pigs but was ineffective in adults (<2.5%). Specific [(3)H]PAF binding was greater in fetus and newborn than in adults; a concordant developmental PAF-induced inositol phosphate formation was observed. PAF-induced vasoconstriction was abrogated by thromboxane A(2) (TXA(2)) synthase and receptor inhibitors, calcium channel blockers, and by removal of endothelium; vasoconstriction to TXA(2) mimetic U-46619 did not differ with age. Immunoreactive TXA(2) synthase expression and PAF-evoked TXA(2) formation revealed a fetus> newborn>adult profile. Thus the greater PAF-induced PV microvascular constriction in younger subjects seems attributable to greater PAF receptor density and mostly secondary to TXA(2) formation from endothelium. The resulting decrease in blood flow may contribute to the increased vulnerability of the PV brain regions to oxidant stress-induced injury in immature subjects.


Subject(s)
Aging/physiology , Brain/blood supply , Brain/drug effects , Platelet Activating Factor/pharmacology , Vasoconstriction/drug effects , Animals , Animals, Newborn , Brain/enzymology , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Fetus/drug effects , Fetus/physiology , Inositol Phosphates/metabolism , Logistic Models , Swine/physiology , Thromboxane B2/metabolism
12.
Invest Ophthalmol Vis Sci ; 43(10): 3327-37, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12356842

ABSTRACT

PURPOSE: To test whether platelet-activating factor (PAF) directly causes retinovascular endothelial cell (EC) death. METHODS: Retinovascular density was calculated in rat pups exposed to 80% O(2) from postnatal days (P)6 to P14 (to produce oxygen-induced retinopathy [OIR]), using the adenosine diphosphatase (ADPase) technique, in animals treated with distinct PAF receptor blockers (PCA-4248, BN52021, or THG315). PAF levels were then measured in the retinas. Viability of ECs from piglets and humans in response to C-PAF (a stable PAF analogue) was determined by the reduction of the tetrazolium salt 3-(4,5-dimethyl thiazol-2yl)-2,5-diphenyl tetrazolium bromide (MTT) by viable cells, incorporation of propidium iodide (PI), TUNEL assay, and release of lactate dehydrogenase. Release of thromboxane (TX) was measured in the cell media. RESULTS: PAF levels in retina were markedly increased by exposure of isolated rat retinas to H(2)O(2) (1 micro M) and of rat pups placed in 80% O(2). Exposure to 80% O(2) induced retinal vasoobliteration, which was equally significantly inhibited ( approximately 60%) by all PAF receptor blockers tested. C-PAF increased incorporation of PI by isolated rat retinal microvasculature. Also, C-PAF caused time- and concentration-dependent death of cultured retinal ECs, which was prevented by the PAF receptor antagonist CV-3988. This effect of C-PAF was selective on retinal and neurovascular ECs, but not on other ECs. DNA fragmentation (TUNEL) was hardly detected, and inhibition of apoptosis-related processes by nicotinamide, cyclosporin A, and Z-DEVD-FMK and Z-VAD-FMK (caspase inhibitors) barely protected against death in EC, whereas C-PAF increased release of lactate dehydrogenase, implying that necrosis is the nature of EC death. Finally, C-PAF-induced cell death was preceded by an increase in TXB(2) levels and was prevented by TXA(2) synthase inhibition (with CGS12970). CONCLUSIONS: The data suggest PAF plays a major role in vasoobliteration in OIR by triggering death of neuroretinal microvascular ECs. The cell death seems to be mediated at least in part by TXA(2). These effects of PAF may participate in ischemic retinopathies such as diabetes and retinopathy of prematurity.


Subject(s)
Oxygen , Platelet Activating Factor/therapeutic use , Receptors, G-Protein-Coupled , Retinal Diseases/chemically induced , Retinal Diseases/drug therapy , Retinal Vessels/drug effects , Animals , Cell Survival/drug effects , Cell Survival/physiology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Humans , Hyperoxia/complications , In Vitro Techniques , Injections , Microcirculation/drug effects , Oxidative Stress , Pericytes/drug effects , Platelet Activating Factor/analogs & derivatives , Platelet Activating Factor/antagonists & inhibitors , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/antagonists & inhibitors , Retinal Diseases/metabolism , Retinal Diseases/pathology , Retinal Vessels/pathology , Retinal Vessels/physiopathology , Swine , Thromboxane A2/physiology , Vitreous Body
SELECTION OF CITATIONS
SEARCH DETAIL
...