Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
Add more filters










Publication year range
1.
Acta Physiol Scand ; 173(1): 23-33, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11678723

ABSTRACT

Ischaemia/reperfusion (I/R) tolerance refers to the phenomenon by which the inflammation and associated sequelae induced by I/R is ameliorated by an I/R challenge imposed 24 h earlier. The development of I/R tolerance is dependent on the synthesis of new proteins. In vivo and in vitro studies provide support for nitric oxide synthase (NOS), antioxidant enzymes, and heat shock proteins (HSPs) as the effector proteins. Activation of the nuclear transcription factor, NFkappaB, appears to be a prerequisite for the development of I/R tolerance. In vitro approaches using anoxia/reoxygenation (A/R) to mimic I/R have provided insights into the complexity of the development of I/R tolerance, i.e. different cells may use different signalling pathways to develop A/R tolerance and influence the responses of adjacent cells during the process. The use of cells from genetically altered mice is expediting attempts to unravel specific mechanisms involved in the development of A/R tolerance.


Subject(s)
Endothelium, Vascular/cytology , Ischemic Preconditioning, Myocardial , Myocardial Reperfusion Injury/pathology , Myocardium/cytology , Animals , Cell Communication/immunology , Endothelium/cytology , In Vitro Techniques
2.
Crit Care Med ; 29(9): 1774-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11546984

ABSTRACT

OBJECTIVE: Peritonitis induced by cecal ligation and perforation results in inflammation and dysfunction of the rat myocardium, an organ remote from the locus of infection. This peritonitis-induced pathology can be prevented by pretreating these animals with lipopolysaccharide before cecal ligation and perforation. In the present study, we assessed a) whether cardiomyocytes obtained from rats subjected to cecal ligation and perforation could induce polymorphonuclear leukocyte transendothelial migration, b) whether these cardiomyocytes could activate endothelial cells (increased proadhesive phenotype), and c) whether these responses could be attenuated by lipopolysaccharide pretreatment. DESIGN: Prospective animal study. SETTING: Experimental animal laboratory. SUBJECTS: Male Sprague Dawley rats. INTERVENTIONS: Lipopolysaccharide pretreated and nonpretreated rats were subjected to cecal ligation and perforation or to laparotomy. Myocytes were isolated 6 hrs after surgery and used for in vitro experiments. MEASUREMENTS AND MAIN RESULTS: Myocytes isolated from cecal ligation and perforation rats promoted migration of polymorphonuclear leukocytes across a rat endothelial cell monolayer, an effect prevented by platelet activating factor receptor antagonists. Myocytes isolated from these animals also increased surface level expression of intercellular adhesion molecule-1 on rat endothelial cells, an effect also prevented by platelet activating factor receptor antagonists. Myocytes isolated from rats pretreated with lipopolysaccharide and then subjected to cecal ligation and perforation did not a) promote polymorphonuclear leukocyte transendothelial migration or b) increase intercellular adhesion molecule-1 surface expression on endothelial cells. CONCLUSIONS: Our findings indicate that induction of peritonitis results in a systemic response that induces cardiac myocytes to become proinflammatory (i.e., these myocytes produce chemotactic factors and activate endothelial cells). This effect of cecal ligation and perforation is abrogated by pretreating animals with lipopolysaccharide before induction of peritonitis.


Subject(s)
Cell Movement , Intercellular Adhesion Molecule-1/pharmacology , Lipopolysaccharides , Myocardium/metabolism , Neutrophils/drug effects , Peritonitis/metabolism , Animals , Endothelium/metabolism , Ligation , Male , Muscle, Skeletal/metabolism , Oxidative Stress , Peritonitis/etiology , Rats , Rats, Sprague-Dawley , Rats, Wistar
3.
Am J Physiol Heart Circ Physiol ; 281(1): H440-7, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11406513

ABSTRACT

The goal of the present study was to assess whether cardiac myocytes exposed to anoxia-reoxygenation (A/R) could generate a chemotactic gradient for polymorphonuclear neutrophil (PMN) transendothelial migration. Exposure of neonatal mouse cardiac myocytes to A/R induced an oxidant stress in the myocytes. Supernatants obtained from A/R-conditioned myocytes promoted mouse PMN migration across mouse myocardial endothelial cell monolayers. This increase in PMN transendothelial migration could be prevented if catalase or a platelet-activating factor (PAF) antagonist was added to the supernatants before assay. Supernatants from A/R-conditioned myocytes activated endothelial cells by inducing an intracellular oxidant stress. The oxidant stress and PMN transendothelial migration induced by supernatants from A/R-conditioned myocytes were substantially reduced when endothelial cells derived from manganese superoxide dismutase overexpressing mice were used in the assays. Supernatants from A/R-conditioned myocytes also increased endothelial cell surface levels of E-selectin and intercellular adhesion molecule-1. Our results indicate that cardiac myocytes exposed to A/R can generate a chemotactic gradient, presumably due to production and release of stable oxidants and PAF. The ability of supernatants from A/R-conditioned myocytes to promote PMN transendothelial migration was largely dependent on induction of an oxidant stress in endothelial cells. In addition, these supernatants also induced a proadhesive phenotype in the endothelial cells.


Subject(s)
Endothelium, Vascular/physiology , Hypoxia/pathology , Hypoxia/physiopathology , Myocardium/pathology , Neutrophils/physiology , Oxygen/pharmacology , Animals , Cell Movement , Cells, Cultured , Mice , Mice, Inbred C57BL
4.
News Physiol Sci ; 16: 15-9, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11390940

ABSTRACT

To reach an inflammatory site in the interstitium, circulating neutrophils (PMN) must first traverse the endothelial barrier. Whether PMN emigrate between endothelial cells (paracellular pathway) or through the endothelial cells proper (transcellular pathway) is controversial. Herein, we present anatomic, functional, and teleological arguments that support both points of view. An attempt is also made to reconcile this apparent controversy.


Subject(s)
Cell Movement/physiology , Endothelium, Vascular/cytology , Neutrophils/cytology , Animals , Humans
5.
Am J Physiol Gastrointest Liver Physiol ; 280(2): G291-7, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11208553

ABSTRACT

In vitro, nitric oxide (NO) decreases leukocyte adhesion to endothelium by attenuating endothelial adhesion molecule expression. In vivo, lipopolysaccharide-induced leukocyte rolling and adhesion was greater in inducible NO synthase (iNOS)-/- mice than in wild-type mice. The objective of this study was to assess E- and P-selectin expression in the microvasculature of iNOS-/- and wild-type mice subjected to acute peritonitis by cecal ligation and perforation (CLP). E- and P-selectin expression were increased in various organs within the peritoneum of wild-type animals after CLP. This CLP-induced upregulation of E- and P-selectin was substantially reduced in iNOS-/- mice. Tissue myeloperoxidase (MPO) activity was increased to a greater extent in the gut of wild-type than in iNOS-/- mice subjected to CLP. In the lung, the reduced expression of E-selectin in iNOS-/- mice was not associated with a decrease in MPO. Our findings indicate that NO derived from iNOS plays an important role in sepsis-induced increase in selectin expression in the systemic and pulmonary circulation. However, in iNOS-/- mice, sepsis-induced leukocyte accumulation is affected in the gut but not in the lungs.


Subject(s)
Bacterial Infections/metabolism , E-Selectin/metabolism , Endothelium, Vascular/metabolism , Nitric Oxide Synthase/deficiency , P-Selectin/metabolism , Acute Disease , Animals , Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout/genetics , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Peritonitis/metabolism , Peritonitis/microbiology , Peroxidase/metabolism , Time Factors , Tissue Distribution , Up-Regulation
6.
Am J Physiol Regul Integr Comp Physiol ; 278(5): R1140-7, 2000 May.
Article in English | MEDLINE | ID: mdl-10801280

ABSTRACT

The objectives of this study were to determine 1) the changes in endothelial cell adhesion molecule expression that occur in a clinically relevant model of sepsis and 2) the dependence of these changes on endotoxin [lipopolysaccharide (LPS)]. The dual radiolabeled monoclonal antibody technique was used to quantify the expression of E- and P-selectin in LPS-sensitive (C3HeB/FeJ) and LPS-insensitive (C3H/HeJ) mice that were subjected to acute peritonitis by cecal ligation and perforation (CLP). At 6 h after CLP, the expression of both E- and P-selectin was increased in the gut (mesentery, pancreas, and small and large bowel) compared with the sham-operated and/or control animals, with a more marked response noted in LPS-insensitive mice. The lung also exhibited an increased P-selectin expression in both mouse strains. An accumulation of granulocytes, assessed using tissue myeloperoxidase activity, was noted in the lung and intestine of LPS-sensitive but not LPS-insensitive mice exposed to CLP. These results indicate that the CLP model of sepsis is associated with an upregulation of endothelial selectins in the gut vasculature and that enteric LPS does not contribute to this endothelial cell activation response.


Subject(s)
E-Selectin/metabolism , Endothelium, Vascular/metabolism , Intestines/blood supply , Lipopolysaccharides/pharmacology , P-Selectin/metabolism , Peritonitis/metabolism , Acute Disease , Animals , Cecum/surgery , Intestinal Mucosa/metabolism , Lactic Acid/blood , Leukocyte Count , Ligation , Male , Mesentery/metabolism , Mice , Mice, Inbred C3H , Neutrophils/pathology , Pancreas/metabolism , Peroxidase/metabolism
7.
Microcirculation ; 7(2): 83-101, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10802851

ABSTRACT

The microvascular dysfunction which occurs in sepsis involves all three elements of the microcirculation: arterioles, capillaries, and venules. In sepsis, the arterioles are hyporesponsive to vasoconstrictors and vasodilators. Sepsis also reduces the number of perfused capillaries, thereby impacting on oxygen diffusion to mitochondria. In the venules of some tissues (e.g., mesentery) there is an inflammatory response characterized by neutrophil infiltration and protein leakage. In addition, PMN-endothelial adhesive interactions occur in precapillary microvessels and capillaries in organs, such as, the lung and heart. Thus, all these elements of the microcirculation are involved in the sepsis-induced inflammation. In this review we address emerging views on the mechanisms involved in the microvascular dysfunction induced by sepsis within the framework of these three basic elements of the microcirculatory unit.


Subject(s)
Inflammation , Microcirculation , Sepsis/physiopathology , Capillary Permeability , Cell Adhesion , Humans , Leukocytes/pathology , Oxygen Consumption , Sepsis/pathology
8.
Am J Physiol Heart Circ Physiol ; 278(3): H853-61, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10710354

ABSTRACT

Cytokine release from inflammatory (CD14(+)) cells is reduced after repeated stimulation with lipopolysaccharide (LPS; LPS tolerance). However, it is not known whether LPS tolerance can be induced in CD14(-) cells. The aim of the present study was to determine whether endothelial cells [human umbilical vein endothelial cells (HUVEC)] could be rendered tolerant to LPS with respect to LPS-induced polymorphonuclear neutrophil (PMN) adhesion. LPS stimulation (0.5 microg/ml; 4 h) of naive HUVEC increased PMN adhesion. Pretreatment of HUVEC with LPS (0.5 microg/ml) for 24 h resulted in a reduction in the proadhesive effects of a subsequent LPS challenge. The initial LPS stimulation increased 1) mobilization of the nuclear transcription factor NF-kappaB to the nucleus and 2) surface levels of the adhesion molecules intercellular adhesion molecule-1 (ICAM-1) and E-selectin. In LPS-tolerant HUVEC, a second LPS challenge resulted in 1) less accumulation of NF-kappaB in the nucleus, 2) a reduction in E-selectin expression, and 3) unchanged ICAM-1 expression. LPS-tolerant cells were still capable of mobilizing NF-kappaB in response to stimulation with either interleukin-1beta or tumor necrosis factor-alpha, resulting in elevated E-selectin levels and increased PMN adhesion. These studies show for the first time that LPS tolerance can be induced in endothelial cells with respect to PMN adhesion. This tolerance is specific for LPS and is associated with an inability of LPS to mobilize NF-kappaB, resulting in less E-selectin expression.


Subject(s)
E-Selectin/metabolism , Endothelium, Vascular/physiology , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Neutrophils/physiology , Cell Adhesion , Drug Tolerance , E-Selectin/analysis , Endothelium, Vascular/chemistry , Humans , Intercellular Adhesion Molecule-1/analysis , Interleukin-1/pharmacology , Lipopolysaccharides/administration & dosage , Tumor Necrosis Factor-alpha/pharmacology , Umbilical Veins
9.
Eur J Pediatr Surg ; 9(4): 196-9, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10532256

ABSTRACT

In summary, with resection of the small bowel, the remaining intestine mounts an adaptive response to increase its absorptive surface area. The malabsorption of nutrients is predicated on the importance of the bowel resected to the absorption of a given nutrient. Motor activity is little effected by small-bowel resection. In general, resection of the upper small intestine is better tolerated than resection of the lower small intestine, particularly if the ileocecal junction is affected.


Subject(s)
Gastrointestinal Motility/physiology , Intestinal Absorption/physiology , Intestine, Small/physiology , Short Bowel Syndrome/physiopathology , Humans
10.
Am J Physiol ; 277(3): H885-92, 1999 09.
Article in English | MEDLINE | ID: mdl-10484407

ABSTRACT

Peritonitis induced by cecal ligation and puncture (CLP) produces a systemic inflammatory response that can be largely mitigated by pretreatment of the animals with lipopolysaccharide (LPS tolerance). Although cells of myeloid origin and endothelial cells have been shown to contribute to the development of LPS tolerance, little is known regarding the potential role of parenchymal cells in this phenomenon. The major aim of the present study was to assess whether cardiac parenchymal cells (myocytes) contribute to the development of LPS tolerance. Six hours after induction of CLP rats were neutropenic and acidotic, the myocardium contained a leukocyte infiltrate [myeloperoxidase (MPO) activity was increased], and myocardial contractile function was impaired (left ventricular developed pressure was decreased). In animals that were pretreated with LPS these manifestations of sepsis were largely reversed. Further studies focused on the responses of cardiac myocytes to CLP and whether myocytes contributed to the development of LPS tolerance. Myocytes were isolated from rat hearts 6 h after induction of CLP. These myocytes 1) exhibited an impaired ability to shorten in response to pacing, 2) contained the nuclear transcription factor NF-kappaB in their nuclei, 3) increased their surface levels of intercellular adhesion molecule-1 (ICAM-1), and 4) were hyperadhesive for neutrophils. All of these events did not occur in myocytes obtained from animals that were pretreated with LPS before induction of CLP. These findings indicate that LPS tolerance can be induced in myocytes with respect to polymorphonuclear leukocyte adhesion, presumably by an inability of CLP to mobilize NF-kappaB to the myocyte nuclei and, thereby, preventing an increase in surface levels of ICAM-1.


Subject(s)
Heart Diseases/physiopathology , Lipopolysaccharides/pharmacology , Myocardium/pathology , Peritonitis/physiopathology , Animals , Cell Adhesion , Heart Diseases/prevention & control , Inflammation/physiopathology , Inflammation/prevention & control , Intercellular Adhesion Molecule-1/physiology , Lipopolysaccharides/therapeutic use , Myocardial Contraction , NF-kappa B/physiology , Neutrophils/pathology , Rats , Rats, Sprague-Dawley
11.
J Cell Sci ; 112 ( Pt 12): 1937-45, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10341212

ABSTRACT

One of the cardinal signs of acute inflammation is neutrophil (PMN) emigration across the endothelium and into the affected tissue. We have previously shown that human PMN migration across human umbilical vein endothelial cell (HUVEC) monolayers is dependent on PMN-derived elastase. However, whether migrating PMN release elastase into the extracellular milieu or retain it on the cell surface is unclear. In the present study, we show that when PMN are activated by platelet activating factor (PAF), elastase was mobilized to and retained in the cell membrane; no elastase activity was detected in the supernatant. Neutroplasts (enucleated cells devoid of granules) prepared from PAF-activated PMN contained twice as much elastase as did neutroplasts prepared from unstimulated PMN. Neutroplasts from PAF-activated PMN migrated across HUVEC monolayers in response to a chemotactic gradient (PAF), while those prepared from unstimulated PMN did not. The neutroplast transendothelial migration was inhibited (80%) by a monoclonal antibody against elastase. Using confocal microscopy, we noted that the localization of elastase on the cell surface of PMN, which were adherent to HUVEC but not migrating, was largely confined to the apical aspect of the PMN. There was little or no elastase detectable on the basal aspect of the PMN membrane in contact with the endothelium. By contrast, in migrating PMN the membrane-bound elastase was primarily localized to the migrating front, i.e. pseudopodia penetrating the HUVEC monolayers. Taken together, our findings indicate that migrating PMN localize their membrane-bound elastase to the migrating front where it facilitates transendothelial migration.


Subject(s)
Endothelium, Vascular/drug effects , Neutrophils/drug effects , Pancreatic Elastase/metabolism , Platelet Activating Factor/pharmacology , Cell Membrane/drug effects , Cell Movement/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Humans , Microscopy, Confocal , Neutrophils/cytology , Neutrophils/enzymology
12.
Circ Res ; 84(1): 103-12, 1999.
Article in English | MEDLINE | ID: mdl-9915779

ABSTRACT

Exposing human umbilical vein endothelial cells (HUVECs) to anoxia/reoxygenation (A/R) results in an increase in polymorphonuclear leukocyte (PMN) adhesion to HUVECs. This A/R-induced hyperadhesion is completely prevented by a previous (24 hours earlier) exposure of HUVECs to A/R. This phenomenon has been termed "A/R tolerance." Exposing HUVECs to A/R induces an increase in nuclear factor kappaB (NF-kappaB) in HUVEC nuclei within 4 hours. Interfering with either NF-kappaB activation (proteasome inhibitor) or translocation (double-stranded oligonucleotides containing NF-kappaB binding sequence) prevents the development of A/R tolerance (ie, the increase in A/R-induced PMN adhesion to HUVECs is the same after the first and second A/R challenges). NO production by HUVECs is increased after the second A/R challenge, but not after the first A/R challenge. Inhibition of NO synthase (NOS) during the second A/R challenge prevents the development of A/R tolerance with respect to PMN adhesion. However, while HUVECs contained endothelial NOS protein, no inducible NOS was detected in either tolerant or nontolerant cells. Further studies indicated that inhibition of GTP-cyclohydrolase I (an enzyme involved in de novo synthesis of an important cofactor for NOS activity, tetrahydrobiopterin) prevented the generation of NO in A/R-tolerant cells. Extracellular generation of NO (NO donor) did not effect the hyperadhesion response induced by the initial A/R challenge. A/R also induced an oxidant stress in naive HUVECs, but not in A/R-tolerant HUVECs. Inhibition of NOS during the second A/R insult results in the generation of an oxidant stress similar to that observed after the first A/R challenge. Taken together, the findings of the present study are consistent with a role for NF-kappaB in the development of A/R tolerance (with respect to PMN adhesion), perhaps by transcriptional regulation of GTP-cyclohydrolase. The increased NO production during the second A/R insult reduces PMN adhesion most likely by reducing the intracellular oxidant stress induced by A/R.


Subject(s)
Cell Adhesion/physiology , Cell Hypoxia , Endothelium, Vascular/physiology , NF-kappa B/metabolism , Neutrophils/physiology , Base Sequence , Binding Sites , Cell Adhesion/drug effects , Cell Nucleus/physiology , Cell Respiration , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Gene Expression Regulation , Humans , Models, Cardiovascular , NF-kappa B/genetics , Neutrophils/drug effects , Nitrates/metabolism , Nitric Oxide/physiology , Nitrites/metabolism , Nitrogen Oxides , Oligodeoxyribonucleotides/metabolism , Oxygen Consumption , Spermine/analogs & derivatives , Spermine/pharmacology , Substrate Specificity , Umbilical Veins
13.
Am J Physiol ; 275(1): H94-9, 1998 07.
Article in English | MEDLINE | ID: mdl-9688900

ABSTRACT

We tested the hypothesis that ischemic preconditioning (PC) of skeletal muscle provided tolerance to a subsequent ischemic event 24 h later, and that such protection was due to nitric oxide (NO). Male Wistar rats, anesthetized with halothane, were randomly assigned to groups: ischemic (no PC; n = 11), PC (n = 11), PC + N-nitro-L-arginine methyl ester (L-NAME; 100 micromol/l; n = 5), PC + N-nitro-D-arginine methyl ester (100 micromol/l; n= 4), PC + aminoguanidine (AMG; 100 micromol/l; n = 4), ischemic + L-NAME (n= 4), or ischemic + AMG (n = 4). PC consisted of 5x 10 min of ischemia and reperfusion, and, 24 h later, 2 h of ischemia were induced by a tourniquet applied to the limb. With the use of intravital microscopy, the number of perfused capillaries (Npc) in the extensor digitorum longus (EDL) muscle was measured over a 90-min reperfusion period. The ratio of ethidium bromide- to bisbenzimide-labeled nuclei was used to estimate tissue injury. PC preserved Npc (23.6 +/- 2.5) following 2 h of ischemia compared with sham muscles (11.5 +/- 5.1), significantly elevating inducible NO synthase (iNOS) activity (81% increase), but did not afford protection to the parenchyma. L-NAME and AMG prevented ischemia-reperfusion-induced reduction in Npc in muscles without PC. However, after 90 min of reperfusion, L-NAME (Npc = 15.0 +/- 1.7), but not AMG (Npc = 22.8 +/- 3.1), significantly reduced the microvascular protection afforded by PC. We conclude that PC of the EDL muscle resulted, 24 h later, in protection to microvascular perfusion only, and that such protection was due to NO from sources other than iNOS.


Subject(s)
Ischemia/prevention & control , Ischemic Preconditioning , Muscle, Skeletal/blood supply , Muscle, Skeletal/physiology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/metabolism , Nitric Oxide/physiology , Animals , Enzyme Inhibitors/pharmacology , Guanidines/pharmacology , Ischemia/physiopathology , Kinetics , Male , Microcirculation/drug effects , Microcirculation/physiology , Microscopy, Video , Muscle, Skeletal/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II , Rats , Rats, Wistar , Stereoisomerism , Time Factors
14.
Circ Res ; 81(6): 922-31, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9400372

ABSTRACT

The objectives of this study were to (1) determine the time course of neutrophil adhesion to monolayers of human umbilical vein endothelial cells (HUVECs) that were exposed to 60 minutes of anoxia followed by 30 to 600 minutes of reoxygenation and (2) define the mechanisms responsible for both the early (minutes) and late (hours) hyperadhesivity of postanoxic HUVECs to human neutrophils. The results clearly demonstrate that anoxia/reoxygenation (A/R) leads to a biphasic increase in neutrophil adhesion to HUVECs, with peak responses occurring at 30 minutes (phase 1) and 240 minutes (phase 2) after reoxygenation. Oxypurinol and catalase inhibited phase-1 adhesion, suggesting a role for xanthine oxidase and H2O2. In comparison, platelet activating factor (PAF) contributed to both phases of neutrophil adhesion. Anti-intercellular adhesion molecule-1 (ICAM-1) and anti-P-selectin antibodies (monoclonal antibodies [mAbs]) attenuated phase-1 neutrophil adhesion, consistent with roles for constitutively expressed ICAM-1 and enhanced surface expression of preformed P-selectin. Phase-2 neutrophil adhesion was attenuated by an anti-E-selectin mAb, indicating a dominant role of this adhesion molecule in the late phase response. Pretreatment with actinomycin D and cycloheximide or with competing ds-oligonucleotides containing the nuclear factor-kappa B or activator protein-1 cognate DNA sequences significantly attenuated phase-2 response, suggesting a role for de novo macromolecule synthesis. Surface expression of ICAM-1, P-selectin, and E-selectin on HUVECs correlated with the phase-1 and -2 neutrophil adhesion responses. Collectively, these findings indicate that A/R elicits a two-phase neutrophil-endothelial cell adhesion response that involves transcription-independent and transcription-dependent surface expression of different endothelial cell adhesion molecules.


Subject(s)
Cell Hypoxia , Endothelium, Vascular/cytology , Neutrophils/physiology , Azepines/pharmacology , Cell Adhesion , Cells, Cultured , Humans , Hydrogen Peroxide/pharmacology , Intercellular Adhesion Molecule-1/physiology , NF-kappa B/metabolism , Triazoles/pharmacology
15.
Circ Res ; 81(4): 618-26, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9314844

ABSTRACT

During an acute inflammatory response polymorphonuclear leukocytes (PMNs) adhere to and emigrate across the venular microvasculature. There is general agreement on the mechanisms involved in PMN adhesive interactions. However, the mechanisms by which PMNs migrate across the endothelial lining remain controversial, particularly with respect to the role of elastase. In the present study, we used human umbilical vein endothelial cells (HUVECs) and PMNs to test the hypothesis that the relative role of PMN-derived elastase may be dependent on the degree of HUVEC retraction within monolayers. A high (10(-7) mol/L), but not a low (10(-10) mol/L), concentration of platelet-activating factor (PAF) caused HUVEC retraction of sufficient magnitude to increase transendothelial protein movement. Elastase inhibitors prevented PMN transendothelial migration in response to the low, but not the high, concentration of PAF. These findings suggest that PMN migration across confluent endothelial cells is elastase dependent, whereas PMN migration across retracted endothelial cells is elastase independent. However, under the latter condition (high concentration of PAF), the two endogenous proteases, alpha 2-macroglobulin and alpha 1-antitrypsin, could interfere with PAF-induced PMN transendothelial migration. Thus, as the concentration of PAF is increased, migrating PMNs use other proteases, in addition to elastase. We also noted that transendothelial protein movement is closely coupled to PMN migration.


Subject(s)
Endopeptidases/physiology , Endothelium, Vascular/cytology , Neutrophils/enzymology , Neutrophils/physiology , Serum Albumin/metabolism , Biological Transport , Cell Movement/drug effects , Cell Movement/physiology , Dose-Response Relationship, Drug , Humans , Leukocyte Elastase/physiology , Neutrophils/drug effects , Osmolar Concentration , Platelet Activating Factor/pharmacology , Protease Inhibitors/pharmacology , alpha 1-Antitrypsin/pharmacology , alpha-Macroglobulins/pharmacology
16.
Inflammation ; 21(3): 313-24, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9246573

ABSTRACT

We observed that normal plasma dramatically reduces neutrophil-endothelial adhesion. Therefore, we identified factors in plasma which might limit PMN adhesion in vitro. We found that the anti-adhesive effect was not mediated by vasoactive lipids present in plasma. Immunoprecipitation of soluble adhesion molecules, P and E-selectins and ICAM-1 restored PMN adhesion to control values. We further examined whether soluble adhesion molecules in plasma might also regulate PMN endothelial migration in response to fMLP (10(-6) M). Plasma significantly reduced PMN migration, and this effect was prevented only by the simultaneous removal of soluble P and E selectins and ICAM-1 together, but not individually. These data show that soluble selectins and ICAM-1 may regulate PMN adhesion and diapedesis, and that alterations in the levels of these molecules may regulate PMN-endothelial interactions in vivo.


Subject(s)
Endothelium, Vascular/physiology , Intercellular Adhesion Molecule-1/physiology , Neutrophils/physiology , Selectins/physiology , Adult , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cells, Cultured , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/physiology , E-Selectin/blood , E-Selectin/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/injuries , Heparin/pharmacology , Humans , In Vitro Techniques , Inflammation Mediators/physiology , Intercellular Adhesion Molecule-1/blood , Lipids/blood , Lipids/pharmacology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/cytology , Neutrophils/drug effects , P-Selectin/blood , P-Selectin/physiology , Selectins/blood , Solubility
17.
Am J Physiol ; 273(6): G1189-99, 1997 12.
Article in English | MEDLINE | ID: mdl-9435543

ABSTRACT

Endothelial cells contribute to a variety of biological responses that facilitate organ function. This critical role of the endothelial cell has resulted in the development of different in vitro models that utilize monolayers of cultured cells to simulate conditions that exist in the intact animal. This review focuses on endothelial cell monolayers as a model system for research on certain pathophysiological conditions affecting the gastrointestinal tract. The advantages and limitations of endothelial cell monolayers are addressed, along with evolving technologies and strategies that hold promise for extending the utility of this in vitro model for studies of gastrointestinal function and disease.


Subject(s)
Endothelium, Vascular/physiology , Endothelium, Vascular/physiopathology , Microcirculation/physiopathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Cell Adhesion Molecules/physiology , Cells, Cultured , Endothelium, Vascular/cytology , Humans , Inflammation , Intestinal Mucosa/pathology , Models, Cardiovascular , Reperfusion Injury
18.
Alcohol Clin Exp Res ; 20(9 Suppl): 347A-349A, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8986235

ABSTRACT

The adherence and emigration of leukocytes have been implicated as a rate-limiting step in the microvascular disturbance in a variety of pathogenic events. The objective of the present study was to investigate leukocyte-endothelial cell adhesion and endothelial barrier function in rat mesenteric microvessels exposed to ethanol, which is known to cause inflammation and injury in various organs. Mesentery of male Wistar rats was used for intravital microscopic observations. Leukocyte adherence and albumin leakage were monitored in single postcapillary venules using the intravital fluorescence microscope. Superfusion of 50 mM ethanol elicited the leukocyte adherence and albumin leakage within 60 min. Pretreatment with a monoclonal antibody directed against either CD18 or intercellular adhesion molecule-1 (ICAM-1) significantly prevented the ethanol-induced increase in leukocyte adherence and decrease in barrier function of endothelium. These results suggest that ethanol-induced leukocyte adherence is mediated by CD18 on leukocytes and ICAM-1 on endothelial cells. The present study further supports that CD18/ ICAM-1-dependent leukocyte-endothelial adhesive interactions lead to macromolecular leakage in the postcapillary venules exposed to ethanol.


Subject(s)
CD18 Antigens/physiology , Capillary Permeability/drug effects , Endothelium, Vascular/drug effects , Intercellular Adhesion Molecule-1/physiology , Leukocyte Adherence Inhibition Test , Mesenteric Veins/drug effects , Animals , Male , Microscopy, Fluorescence , Rats , Rats, Wistar , Serum Albumin/metabolism , Venules/drug effects
19.
Gastroenterology ; 110(1): 21-9, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8536858

ABSTRACT

BACKGROUND & AIMS: Previous studies indicate that a water extract of Helicobacter pylori promotes leukocyte adhesion and emigration as well as endothelial barrier disruption (increased vascular protein leakage) in rat mesenteric venules. The aims of this study were to assess whether H. pylori extract-activated neutrophils disrupt endothelial cell monolayers and to identify the mechanisms involved in this process. METHODS: Human neutrophils were incubated with monolayers of human umbilical vein endothelial cells (HUVECs) in the presence or absence of H. pylori extract. RESULTS: H. pylori extract-activated human neutrophils produced endothelial cell detachment from HUVEC monolayers, the severity of which was dependent on the duration of exposure. Endothelial cell detachment was prevented by a monoclonal antibody directed against CD11/CD18 on neutrophils or a monoclonal antibody against intercellular adhesion molecule 1 on endothelial cells. HUVEC monolayer disruption was also prevented by superoxide dismutase, catalase, and a monoclonal antibody against elastase. Further studies indicated that H. pylori extract was capable of inhibiting human neutrophil elastase. The antielastase activity was not diminished by oxidants. CONCLUSIONS: These studies indicate that H. pylori extract-activated human neutrophils can disrupt HUVEC monolayers only when human neutrophils are allowed to adhere to HUVECs and may provide an explanation for the H. pylori extract-induced, neutrophil-dependent vascular protein leakage observed in vivo. The possibility that H. pylori releases antiproteases may explain, in part, why this bacterium is so virulent.


Subject(s)
Endothelium, Vascular/pathology , Helicobacter pylori/metabolism , Leukocyte Elastase/antagonists & inhibitors , Neutrophils/physiology , Pancreatic Elastase/antagonists & inhibitors , Antibodies, Monoclonal , Catalase/pharmacology , Cell Adhesion/drug effects , Cells, Cultured , Culture Media , Humans , Hypochlorous Acid/pharmacology , Superoxide Dismutase/pharmacology
20.
Am J Physiol ; 269(6 Pt 2): H1955-64, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8594904

ABSTRACT

The aim of the present study was to characterize and compare the expression of intercellular adhesion molecule 1 (ICAM-1) on unstimulated and endotoxin-challenged endothelial cells in different tissues of the rat. ICAM-1 expression was measured using 125I-labeled anti-rat ICAM-1 monoclonal antibody (MAb) and an isotype-matched control MAb labeled with 131I (to correct for nonspecific accumulation of the binding MAb). Under baseline conditions, ICAM-1 MAb binding was observed in all organs. The binding of 125I-ICAM-1 MAb varied widely among organs, with the largest accumulation (per g tissue) in the lung, followed by heart (1/30th of lung activity), splanchnic organs (1/50th of lung activity), thymus (1/100th of lung activity), testes (1/300th of lung activity), and skeletal muscle (1/800th of lung activity). Endotoxin induced an increase in ICAM-1 MAb binding in all organs except the spleen. Endotoxin-induced upregulation of ICAM-1 was greatest in heart and skeletal muscle (5- to 10-fold), whereas the remaining organs exhibited a two- to fourfold increase in ICAM-1 expression. Maximal upregulation of ICAM-1 occurred at 9-12 h after endotoxin administration. A dose-dependent increase in ICAM-1 expression was elicited by 0.1-10 microgram/kg, with higher doses (up to 5 mg/kg) producing no further increment. Induction of ICAM-1 mRNA after endotoxin was observed in all tissues examined (lung, heart, intestine), peaked at 3 h, and then rapidly returned to control levels. These findings indicate that ICAM-1 is constitutively expressed on vascular endothelium in all organs of the rat and that there are significant regional differences in the magnitude and time course of endotoxin-induced ICAM-1 expression.


Subject(s)
Intercellular Adhesion Molecule-1/metabolism , Animals , Antibodies, Monoclonal , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Endotoxins/pharmacology , Intercellular Adhesion Molecule-1/genetics , Iodine Radioisotopes , Male , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...