Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Exp Med ; 165(5): 1393-402, 1987 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-3572302

RESUMEN

Selective deacylation of the nonhydroxylated fatty acids from S. typhimurium LPS by an acyloxyacyl hydrolase isolated from leukocytes reduces toxic activity of LPS in vivo. We examined the effect of deacylated LPS on neutrophil adherence to human umbilical vein endothelial cells (HUVE). Pretreatment of HUVE with LPS (13 ng/ml for 4 h) produced a marked increase in the adherence of subsequently added neutrophils. In contrast, there was no increase in the adherence of neutrophils to HUVE pretreated with deacylated LPS (up to 260 ng/ml for 4 h). Neutrophil adherence to HUVE pretreated with LPS decreased as the degree of LPS deacylation increased. Deacylated LPS was not only itself inactive, but it inhibited neutrophil-endothelial interactions induced by LPS. Neutrophil adherence to HUVE pretreated with LPS was inhibited by deacylated LPS in a dose-dependent manner. Complete inhibition of adherence was observed at a 20:1 ratio (wt/wt) of deacylated LPS to LPS. Significantly, inhibition of neutrophil adherence to HUVE pretreated with LPS was observed even when deacylated LPS was added to HUVE up to 60 min after LPS. Deacylated LPS, however, did not inhibit neutrophil adherence induced by pretreatment of HUVE with IL-1 or TNF-alpha. We conclude that enzymatic deacylation of the nonhydroxylated fatty acids of LPS abolishes the ability of LPS to induce surface expression of a neutrophil adherence promoting activity in HUVE. Furthermore, deacylated LPS inhibits neutrophil adherence to HUVE induced by LPS, perhaps by preventing the interaction of LPS with a specific cell-surface or intracellular target.


Asunto(s)
Endotelio/efectos de los fármacos , Lipopolisacáridos/farmacología , Neutrófilos/citología , Adhesión Celular/efectos de los fármacos , Humanos , Lipopolisacáridos/antagonistas & inhibidores
2.
J Leukoc Biol ; 52(6): 679-86, 1992 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-1464739

RESUMEN

Macrophages contribute to the systemic inflammatory response that characterizes the sepsis syndrome through the production of inflammatory cytokines such as tumor necrosis factor (TNF). Liposome-encapsulated hemoglobin (LEH), a potential red cell substitute, is cleared by fixed tissue macrophages. In these studies, in vitro incubation of alveolar macrophages with stored LEH was shown to inhibit the expression of TNF induced by endotoxin (lipopolysaccharide, LPS) stimulation. This effect was dependent on LEH dose but independent of the period of exposure to the LEH. Despite inhibition of TNF expression, Northern blot analysis of total cellular RNA from LPS-stimulated macrophages revealed accumulations of TNF-specific transcripts in cells treated with or without LEH. Thus the mechanism of LEH inhibition of TNF expression appears to involve a posttranscriptional event. Although these results suggest a potential advantage of resuscitation with LEH when sepsis complicates hemorrhagic shock, immunomodulation in vivo remains to be defined.


Asunto(s)
Hemoglobinas/farmacología , Macrófagos Alveolares/fisiología , Procesamiento Postranscripcional del ARN , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Northern Blotting , Células Cultivadas , Portadores de Fármacos , Cinética , Lipopolisacáridos/farmacología , Liposomas , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/ultraestructura , Microscopía Electrónica , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Conejos , Factores de Tiempo , Factor de Necrosis Tumoral alfa/genética
3.
FEBS Lett ; 331(3): 285-90, 1993 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-7690717

RESUMEN

The role of protein kinase C (PKC) in interleukin-1 beta- (II-1 beta)-, tumor necrosis factor-alpha- (TNF-alpha)-, and lipopolysaccharide- (LPS)-induced vascular cell adhesion molecule-1 (VCAM-1) expression on human umbilical vein endothelial cells (HUVEC) was studied. PKC inhibition or downregulation diminished VCAM-1 mRNA accumulation and protein expression. Interleukin-1 beta, TNF-alpha, and LPS induce nuclear factor (NF)-kappa B-like binding activity, which precedes VCAM-1 transcription. PKC inhibition did not prevent NF-kappa B-like binding activity, indicating that this is PKC-independent, and NF-kappa B-like binding activity is insufficient for transcription of VCAM-1.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Endotelio Vascular/metabolismo , Proteína Quinasa C/fisiología , Secuencia de Bases , Moléculas de Adhesión Celular/genética , Membrana Celular/metabolismo , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Selectina E , Activación Enzimática , Expresión Génica/efectos de los fármacos , Humanos , Técnicas In Vitro , Interleucina-1/farmacología , Lipopolisacáridos/farmacología , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Oligodesoxirribonucleótidos/química , Proteína Quinasa C/antagonistas & inhibidores , ARN Mensajero/genética , Acetato de Tetradecanoilforbol/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Venas Umbilicales , Molécula 1 de Adhesión Celular Vascular
4.
J Thorac Cardiovasc Surg ; 116(1): 114-21, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9671905

RESUMEN

INTRODUCTION: Interleukin-8 is thought to play a role in neutrophil activation and transcapillary migration into the interstitium. Because neutrophils are principal effector cells in acute myocardial ischemia-reperfusion injury, we postulated that the inhibition of interleukin-8 activity with a neutralizing monoclonal antibody directed against rabbit interleukin-8 (ARIL8.2) would attenuate the degree of myocardial injury encountered during reperfusion. METHODS: In New Zealand White rabbits, the large branch of the marginal coronary artery supplying most of the left ventricle was occluded for 45 minutes, followed by 2 hours of reperfusion. Fifteen minutes before reperfusion, animals were given an intravenous bolus of either 2 mg/kg of ARIL8.2 or 2 mg/kg anti-glycoprotein-120, an isotype control antibody that does not recognize interleukin-8. At the completion of the 120-minute reperfusion period, infarct size was determined. RESULTS: In the area at risk for infarction, 44.3% +/- 4% of the myocardium was infarcted in the anti-glycoprotein-120 group compared with 24.8% +/- 9% in the ARIL8.2 group (p < 0.005). In control animals, edema and diffuse infiltration of neutrophils were observed predominantly in the infarct zone and the surrounding area at risk. Tissue myeloperoxidase determinations did not differ significantly between groups, indicating that the cardioprotective effect of ARIL8.2 was independent of an effect on neutrophil infiltration. CONCLUSIONS: A specific monoclonal antibody that neutralizes interleukin-8 significantly reduces the degree of necrosis in a rabbit model of myocardial ischemia-reperfusion injury.


Asunto(s)
Interleucina-8/antagonistas & inhibidores , Daño por Reperfusión Miocárdica/prevención & control , Animales , Anticuerpos Monoclonales/farmacología , Presión Sanguínea/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Interleucina-8/sangre , Interleucina-8/inmunología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Activación Neutrófila/efectos de los fármacos , Activación Neutrófila/fisiología , Neutrófilos/fisiología , Peroxidasa/metabolismo , Conejos , Flujo Sanguíneo Regional/efectos de los fármacos
5.
J Thorac Cardiovasc Surg ; 118(1): 154-62, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10384198

RESUMEN

BACKGROUND: The transcription factor nuclear factor kappaB mediates the expression of a number of inflammatory genes involved in the whole-body inflammatory response to injury. We and others have found that dithiocarbamates specifically inhibit nuclear factor kappaB-mediated transcriptional activation in vitro. OBJECTIVE: We hypothesized that inhibition of nuclear factor kappaB with dithiocarbamate treatment in vivo would attenuate interleukin 1 alpha-mediated hypotension in a rabbit model of systemic inflammation. METHODS: New Zealand White rabbits were anesthetized and cannulated for continuous hemodynamic monitoring during 240 minutes. Rabbits were treated intravenously with either phosphate-buffered saline solution or 15 mg/kg of a dithiocarbamate, either pyrrolidine dithiocarbamate or proline dithiocarbamate, 60 minutes before the intravenous infusion of 5 micrograms/kg interleukin 1 alpha. Nuclear factor kappaB activation was evaluated by electrophoretic gel mobility shift assay of whole-tissue homogenates. RESULTS: Infusion of interleukin 1 alpha resulted in significant decreases in mean arterial pressure and systemic vascular resistance, both of which were prevented by treatment with dithiocarbamate. Pyrrolidine dithiocarbamate induced a significant metabolic acidosis, whereas proline dithiocarbamate did not. Nuclear factor kappaB-binding activity was increased within heart, lung, and liver tissue 4 hours after interleukin 1 alpha infusion. Treatment with dithiocarbamate resulted in decreased nuclear factor kappaB activation in lung and liver tissue with respect to that in control animals. CONCLUSIONS: These results demonstrate that nuclear factor kappaB is systemically activated during whole-body inflammation and that inhibition of nuclear factor kappaB in vivo attenuates interleukin 1 alpha-induced hypotension. Nuclear factor kappaB thus represents a potential therapeutic target in the treatment of hemodynamic instability associated with the whole-body inflammatory response.


Asunto(s)
Antioxidantes/uso terapéutico , Hemodinámica/efectos de los fármacos , Hipotensión/etiología , Hipotensión/fisiopatología , Interleucina-1/efectos adversos , FN-kappa B/efectos de los fármacos , FN-kappa B/inmunología , Prolina/análogos & derivados , Pirrolidinas/uso terapéutico , Síndrome de Respuesta Inflamatoria Sistémica/complicaciones , Síndrome de Respuesta Inflamatoria Sistémica/tratamiento farmacológico , Tiocarbamatos/uso terapéutico , Acidosis/inducido químicamente , Animales , Antioxidantes/farmacología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Infusiones Intravenosas , Interleucina-1/administración & dosificación , Interleucina-1/inmunología , FN-kappa B/análisis , Prolina/farmacología , Prolina/uso terapéutico , Pirrolidinas/farmacología , Conejos , Distribución Aleatoria , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Tiocarbamatos/farmacología
6.
Surgery ; 106(2): 216-22; discussion 222-3, 1989 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-2669197

RESUMEN

Human endothelial cells exposed to lipopolysaccharide (LPS), tumor necrosis factor (TNF), or interleukin-1 (IL-1) in vitro acquire a cell surface property that promotes the adherence of neutrophils (PMNs). The common mechanism by which endothelial cells are activated by these agents is unknown. We examined adherence of PMNs to cultured human umbilical vein endothelium (HUVE) pretreated with LPS (100 ng/ml), TNF (100 U/ml), and IL-1 (1 U/ml) in medium alone or medium containing protein kinase inhibitors H-7 or HA-1004. Both compounds inhibit a similar spectrum of protein kinases, but H-7 is an effective inhibitor of protein kinase C, whereas HA-1004 is not. We found that H-7 (25 mumol/L) reduced the adherence of PMNs to LPS-, TNF-, and IL-1-stimulated HUVE monolayers to 16.7% +/- 3.0%, 12.1% +/- 2.5%, and 18.3% +/- 2.9% of control, respectively (mean plus or minus standard error of three experiments); HA-1004 (25 mumol/L) did not inhibit endothelial adhesiveness. Cytotoxicity of H-7 was less than 10% in LPS-, TNF-, and IL-1-treated HUVE. Protein synthesis, as measured by the incorporation of tritiated amino acids, was not significantly impaired in LPS-treated HUVE concurrently exposed to H-7. We conclude that protein kinase C appears to be a necessary common mediator of endothelial cell activation by LPS, TNF, and IL-1.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Endotoxinas/farmacología , Escherichia coli , Interleucina-1/farmacología , Proteína Quinasa C/fisiología , Factor de Necrosis Tumoral alfa/farmacología , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina , Adhesión Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Humanos , Isoquinolinas/farmacología , Neutrófilos/fisiología , Piperazinas/farmacología , Biosíntesis de Proteínas , Inhibidores de Proteínas Quinasas , Proteínas Recombinantes
7.
Surgery ; 106(2): 339-45; discussion 345-6, 1989 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-2788318

RESUMEN

Interleukin-2 (IL-2) therapy for patients with advanced cancer may be compromised by dose-limiting and life-threatening pulmonary and systemic edema. We studied the effects of bolus IL-2 infusion on lung and soft-tissue transvascular fluid and protein filtration in six sheep with chronic lung and soft-tissue lymphatic cannulation. Changes in lung (QL) and soft-tissue (QS) lymph flow were used as indicators of transvascular fluid filtration. A dose of 100,000 U/kg IL-2 was administered every 8 hours for 3 days. A significant increase (p less than or equal to 0.05) in both QL and QS was observed after each IL-2 infusion, with maximal flow occurring 2 to 3 hours after infusion. After 72 hours of IL-2 infusion, a fourfold maximal increase in QL occurred, which recovered to near-baseline values within 24 hours. Elevations in QL and QS were not associated with increases in pulmonary arterial or pulmonary arterial wedge pressures, but these elevations were associated with significant (p less than or equal to 0.05) increases in cardiac output (7.7 +/- 0.5 to 11.4 +/- 0.4 L/min) and a consistent decrease in systemic vascular resistance. A significant increase in lung lymph/plasma protein ratio (0.49 +/- 0.06 to 0.93 +/- 0.04 for albumin) revealed a marked increase in pulmonary microvascular porosity. This change, however, was not observed in the systemic microcirculation. Serum concentrations of tumor necrosis factor did not increase with the observed changes in pulmonary microvascular porosity. We conclude that IL-2 increases both pulmonary and systemic microvascular fluid flux. In addition, there is a marked increase in pulmonary, but not systemic, protein permeability that is not a consequence of changes mediated by tumor necrosis factor.


Asunto(s)
Vasos Sanguíneos/metabolismo , Líquidos Corporales/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Interleucina-2/farmacología , Circulación Pulmonar , Animales , Pulmón/metabolismo , Linfa/metabolismo , Proteínas/metabolismo , Ovinos , Factor de Necrosis Tumoral alfa/farmacología , Resistencia Vascular/efectos de los fármacos
8.
Surgery ; 108(2): 206-12, 1990 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-1974365

RESUMEN

Neutrophil adherence or aggregation may be important in the development of organ injury after hemorrhagic shock. Monoclonal antibody (MAb) 60.3 prevents both adherence and aggregation. Therefore we investigated MAb 60.3 treatment in prevention of organ injury after hemorrhagic shock in rhesus monkeys (Macaca mulatta). We performed esophagogastroscopy and placed catheters to measure cardiac output, mean arterial pressure, arterial blood gases, and urine output. Blood was removed to decrease CO to 30% of baseline for 90 minutes. Just before resuscitation, MAb 60.3 (2 mg/kg) or saline solution (control) was administered intravenously. Monitoring and fluid resuscitation continued for 24 hours, with lactated Ringer's solution given as a maintenance infusion (4 ml/kg/hr) plus additional lactated Ringer's solution to maintain CO at preshock levels. Esophagogastroscopy was repeated 24 hours after shock. There were two deaths in the control group at about 72 hours and none in the MAb 60.3 group. MAb 60.3-treated animals required less fluid (9.6 +/- 8.8 ml/kg vs 263.8 +/- 225.7 ml/kg), gained less weight (0.08 +/- 0.11 kg vs 0.70 +/- 0.37 kg), and maintained a higher hematocrit level (35.0% +/- 1.0% vs 26.9% +/- 4.9%). All five control animals had gastritis; MAb 60.3-treated animals had none (p less than 0.05; Fisher's exact test). Inhibition of neutrophil adherence or aggregation with MAb 60.3 at the time of resuscitation reduces fluid requirements and gastric injury in monkeys after hemorrhagic shock.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Insuficiencia Multiorgánica/prevención & control , Neutrófilos/efectos de los fármacos , Receptores de Adhesión de Leucocito/inmunología , Choque Hemorrágico/complicaciones , Animales , Anticuerpos Monoclonales/uso terapéutico , Antígenos CD/inmunología , Arterias , Recuento de Células Sanguíneas , Antígenos CD18 , Gasto Cardíaco , Adhesión Celular/efectos de los fármacos , Fluidoterapia , Gastritis/etiología , Macaca mulatta , Insuficiencia Multiorgánica/inmunología , Neutrófilos/inmunología , Oxígeno/sangre , Presión Parcial , Choque Hemorrágico/patología , Choque Hemorrágico/terapia
9.
Ann Thorac Surg ; 63(1): 277-84, 1997 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8993292

RESUMEN

Many of the components currently used to perform cardiovascular operations lead to systemic insults that result from cardiopulmonary bypass circuit-induced contact activation, circulatory shock, and resuscitation, and a syndrome similar to endotoxemia. Experimental observations have demonstrated that these events have profound effects on activating endothelial cells to recruit neutrophils from the circulation. Once adherent to the endothelium, neutrophils release cytotoxic proteases and oxygen-derived free radicals, which are responsible for much of the end-organ damage seen after cardiovascular operations. Recently the cellular and molecular mechanisms of endothelial cell activation have become increasingly understood. It is conceivable that once the molecular mechanisms of endothelial cell activation are better defined, therapies will be developed allowing the selective or collective inhibition of vascular endothelial activation during the perioperative period.


Asunto(s)
Procedimientos Quirúrgicos Cardíacos , Puente Cardiopulmonar , Endotelio Vascular , Complicaciones Posoperatorias , Síndrome de Respuesta Inflamatoria Sistémica , Procedimientos Quirúrgicos Vasculares , Moléculas de Adhesión Celular/metabolismo , Activación de Complemento , Endotelio Vascular/inmunología , Endotelio Vascular/lesiones , Endotelio Vascular/metabolismo , Humanos , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/inmunología , Síndrome de Respuesta Inflamatoria Sistémica/etiología , Síndrome de Respuesta Inflamatoria Sistémica/inmunología
10.
Ann Thorac Surg ; 62(6): 1868-75, 1996 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8957415

RESUMEN

Myocardial ischemia and reperfusion is a common occurrence in cardiovascular surgery patients. Acute ischemia results in a spectrum of derangements, which range from transient reversible stunning of the myocardium to severe irreversible abnormalities such as infarction. Many of these abnormalities are accentuated upon reperfusion with oxygenated blood. Recently, the endothelium has been shown to play a key role in the injury suffered after ischemia and reperfusion. When rendered hypoxic and then reoxygenated, endothelial cells become activated to express proinflammatory properties that include the induction of leukocyte-adhesion molecules, procoagulant factors and vasoconstrictive agents that increase vasomotor tone. These changes may contribute to the no-reflow phenomenon by promoting endothelial edema, neutrophil and platelet plugging, microthrombosis, and enhanced vasomotor tone. An increased understanding of the role that hypoxic endothelial cell activation plays in myocardial dysfunction after ischemia/reperfusion may allow therapies to be designed to further attenuate this response.


Asunto(s)
Procedimientos Quirúrgicos Cardíacos/efectos adversos , Endotelio Vascular/fisiopatología , Daño por Reperfusión Miocárdica/fisiopatología , Puente Cardiopulmonar/efectos adversos , Humanos , Leucocitos/fisiología , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/terapia , Neutrófilos/fisiología
11.
Ann Thorac Surg ; 68(2): 377-82, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10475400

RESUMEN

BACKGROUND: Ischemic preconditioning (IP) is the phenomenon whereby brief episodes of ischemia protect the heart against a subsequent ischemic stress. We hypothesize that activation of the transcription factor NF-kappaB mediates IP. METHODS: Rabbits were randomly allocated to one of three groups: (1) 45 minutes of myocardial ischemia followed by 2 hours of reperfusion (I/R); (2) three cycles of 5-minute ischemia and 5 minutes of reperfusion followed by I/R (IP + I/R); or (3) IP in the presence of ProDTC, a specific NF-kappaB inhibitor, followed by I/R (IPProDTC + I/R). Infarct size, indices of regional contractility, and NF-kappaB activation were determined. RESULTS: In preconditioned rabbits (IP + I/R), infarct size was reduced 83% compared with both I/R alone and IPProDTC + I/R groups (p < 0.05). Throughout reperfusion, preconditioned myocardium showed enhanced regional contractile function compared with I/R and IPProDTC + I/R groups (p < 0.05). Gel shift analysis showed NF-kappaB activation with IP that was blocked by ProDTC. I/R and IPProDTC + I/R groups showed NF-kappaB activation with I/R that was absent in preconditioned animals. CONCLUSIONS: The cytoprotective effects induced by IP require activation of NF-kappaB.


Asunto(s)
Precondicionamiento Isquémico Miocárdico , Isquemia Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/fisiopatología , FN-kappa B/fisiología , Animales , Electroforesis en Gel de Poliacrilamida , Contracción Miocárdica/fisiología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Isquemia Miocárdica/patología , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Conejos
12.
Ann Thorac Surg ; 68(5): 1949-53, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10585109

RESUMEN

Exacerbation of, rather than improvement in, a hypoxic injury after reperfusion of ischemic tissues is recognized as the specific clinicopathologic entity referred to as ischemia/reperfusion (I/R) injury. Arguably, one of the most common forms of I/R injury occurs during cardiac surgery, which has a mandatory period of myocardial ischemia required to allow surgery in a bloodless, motionless field, followed by coronary artery reperfusion after removal of the aortic cross-clamp. In this review, we examine the endothelial cell activation phenotype that initiates and propagates myocardial I/R injury. Emphasis is given to the biology of one transcription factor, NF-kappaB, that has the principal role in the regulation of many endothelial cell genes expressed in activated endothelium. NF-kappaB-dependent transcription of endothelial cell genes that are transcribed in response to I/R injury may be a favorable approach to preventing tissue injury in the setting of I/R. Elucidating safe and effective therapy to inhibit transcription of endothelial cell genes involved in promoting injury after I/R injury may have wide applicability to the patients with heart disease and other forms of I/R injury.


Asunto(s)
Endotelio Vascular/fisiopatología , Daño por Reperfusión Miocárdica/genética , FN-kappa B/fisiología , Transcripción Genética/genética , Animales , Regulación de la Expresión Génica/fisiología , Humanos , Daño por Reperfusión Miocárdica/fisiopatología , Estrés Oxidativo/genética , Síndrome de Respuesta Inflamatoria Sistémica/genética , Trombomodulina/genética
13.
J Surg Res ; 89(1): 85-119, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10720457

RESUMEN

It is now established that endothelial cells acquire several functional properties in response to a diverse array of extracellular stimuli. This expression of an altered phenotype is referred to as endothelial cell activation, and it includes several activities that promote inflammation and coagulation. While it is recognized that endothelial cell activation has a principal role in host defense, recent studies also demonstrate that endothelial cells are capable of complex molecular responses that protect the endothelium against various forms of stress including heat shock, hypoxia, oxidative stress, shock, ischemia-reperfusion injury, toxins, wounds, and mechanical stress. In this review, we examine endothelial cell genotypic and phenotypic responses to stress. Also, we highlight important cellular stress responses that, although not yet demonstrated directly in endothelial cells, likely exist as part of the repertoire of stress responses in endothelium. A detailed understanding of the molecular mechanisms mediating the adaptive responses of endothelial cells to stress should facilitate the development of novel therapeutics to aid in the management of diverse surgical diseases and their complications.


Asunto(s)
Adaptación Fisiológica , Endotelio Vascular/fisiopatología , Estrés Fisiológico/fisiopatología , Animales , Apoptosis/fisiología , Endotelio Vascular/patología , Endotelio Vascular/fisiología , Humanos , Hipoxia/fisiopatología , Precondicionamiento Isquémico , Neovascularización Patológica/fisiopatología , Estrés Oxidativo/fisiología , Estrés Mecánico , Estrés Fisiológico/patología
14.
Cell Immunol ; 119(1): 41-52, 1989 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-2784080

RESUMEN

In this study we assessed the viability of cultured human umbilical vein endothelial cells (HUVE) treated with bacterial lipopolysaccharide (LPS), recombinant human interleukin-1 (rhIL-1), or recombinant human tumor necrosis factor-alpha (rhTNF-alpha) during inhibition of RNA or protein synthesis. Cytotoxicity was determined by 51Cr activity retained in labeled HUVE monolayers after exposure to LPS, rhIL-1 or rhTNF-alpha, and cycloheximide (Cx) or actinomycin D (Act D). Lipopolysaccharide (150 ng/ml), rhIL-1 (100 pg/ml), or rhTNF-alpha (20 ng/ml) alone was not toxic to HUVE in an 18-hr incubation. Cycloheximide alone (1 microgram/ml for 18 hr) or Act D alone (1 microgram/ml for 6 hr) was also not toxic to HUVE. However, coincubation of HUVE with Cx and LPS (150 ng/ml), rhIL-1 (10 pg/ml), or rhTNF-alpha (20 ng/ml) produced significant cytotoxicity at 18 hr (70 +/- 4% for LPS, 75 +/- 5% for rhIL-1, and 52 +/- 5% for rhTNF-alpha; mean +/- SEM of 18, 16, and 19 separate experiments, respectively). Similarly, coincubation of HUVE with Act D and LPS, rhIL-1, or rhTNF-alpha resulted in 82 +/- 5%, 85 +/- 3%, and 67 +/- 4% cytotoxicity, respectively, at 6 hr (mean +/- SEM of 5 separate experiments for LPS, and 7 separate experiments each for rhIL-1 and rhTNF-alpha). At the highest concentrations of LPS, rhIL-1, or rhTNF-alpha, cytotoxicity during coincubation with Cx or Act D was detected as early as 2 hr and was near maximal by 6 hr. In contrast to LPS, rhIL-1, or rhTNF-alpha, recombinant human interferon-gamma (up to 100 U/ml), or human alpha-thrombin (up to 10 U/ml), produced no cytotoxicity in the presence of Cx. Recombinant human lymphotoxin (up to 50 ng/ml) had a detectable cytotoxic effect in the presence of Cx although it was significantly less than that seen with rhTNF-alpha. Furthermore, coincubation of human fibroblasts and human smooth muscle cells with Cx and LPS, rhIL-1, or rhTNF-alpha produced no cytotoxicity. We conclude that under these culture conditions, LPS, rhIL-1, or rhTNF-alpha produces a lethal injury to HUVE when de novo RNA or protein synthesis is inhibited. These results suggest that LPS, rhIL-1, and rhTNF-alpha may act via a common pathway in endothelial cells and that protein synthesis is important in regulating the response to these stimuli.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Interleucina-1/farmacología , Lipopolisacáridos/farmacología , Biosíntesis de Proteínas , Factor de Necrosis Tumoral alfa/farmacología , Células Cultivadas , Cicloheximida/farmacología , Dactinomicina/farmacología , Endotelio Vascular/metabolismo , Humanos , Proteínas Recombinantes/farmacología
15.
Toxicol Appl Pharmacol ; 124(1): 1-9, 1994 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-7507267

RESUMEN

In view of the potential role of the cytochrome P450 (CYP) and microsomal epoxide hydrolase (mEH) biotransformation enzymes in the metabolism of protoxicants in the circulatory system, we examined CYP and mEH expression in several primary cultures of human umbilical vein endothelial cells (HUVEC), each established from a different individual. Total RNA was isolated from untreated cells and cells 72 hr after exposure to dimethyl sulfoxide (DMSO), Arochlor 1254 (PCB), and beta-naphthoflavone (beta NF). Specific mRNA transcripts were examined by Northern blotting and reverse transcriptase-coupled polymerase chain reaction (RT/PCR) analyses. CYP2E1, CYP3A, and CYP1A2 mRNAs were not detectable in any of the cultures by Northern blot analysis with radiolabeled oligomer probes; however, CYP1A1 mRNA was detected using this procedure in HUVEC cultures exposed to beta NF for 72 hr. Using RT/PCR, constitutive levels of CYP1A1, CYP1A2, CYP2E1, and CYP3A gene expression in HUVEC cultures were evident; however, constitutive CYP2B6 mRNA was not detected. Constitutive CYP1A2 transcript levels were detected in four of six HUVEC cultures, but levels varied between individual cultures. CYP1A2 mRNA levels were also increased in HUVEC cultures exposed to PCB and beta NF. No increases in the levels of CYP2E1 and CYP3A mRNAs were observed in HUVEC cells subsequent to PCB or beta NF exposures. Constitutive CYP2E1 transcript levels were present in all HUVEC cultures examined and varied among individuals. All HUVEC cultures examined for mEH activity exhibited constitutive levels of mEH which varied 40% between individual cultures and produced on average, 1.51 pmol benzo[a]pyrene 4,5-dihydrodiol per milligram protein per minute of reaction. Thus, these results demonstrate that human endothelial cells express CYP and mEH gene products and suggest that these enzymes may play important roles in determining metabolic fates for circulating protoxicants.


Asunto(s)
Sistema Enzimático del Citocromo P-450/genética , Endotelio Vascular/enzimología , Epóxido Hidrolasas/genética , Secuencia de Bases , Northern Blotting , Células Cultivadas , Cromatografía Líquida de Alta Presión , Citocromo P-450 CYP2E1 , Sistema Enzimático del Citocromo P-450/análisis , Sistema Enzimático del Citocromo P-450/fisiología , Endotelio Vascular/fisiología , Epóxido Hidrolasas/fisiología , Expresión Génica , Humanos , Datos de Secuencia Molecular , Oxidorreductasas N-Desmetilantes/análisis , Reacción en Cadena de la Polimerasa , ARN/análisis , Venas Umbilicales/enzimología , Venas Umbilicales/fisiología
16.
J Trauma ; 30(12 Suppl): S49-57, 1990 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-2254991

RESUMEN

Acute pulmonary failure or ARDS in severely injured patients continues to be a significant problem. The most important clinical risk factor identified is sepsis syndrome. Sepsis syndrome is the clinical correlate of a malignant systemic inflammatory process and is directed in large part by the tissue-fixed macrophage (M phi), such as the alveolar M phi. The M phi is capable of producing most of the central inflammatory mediators responsible for the pathophysiology seen during sepsis and organ injury. Two major mediators are procoagulant activity (PCA), leading to diffuse microvascular thrombosis, and tumor necrosis factor (TNF), causing much of the physiologic derangement of sepsis. Endotoxins (LPS) derived from Gram-negative bacterial cell walls are the primary inflammatory stimulus for the tissue-fixed M phi production of inflammatory mediators. It is not completely known how LPS interacts with its various cellular targets, but it is hoped that knowledge of the molecular interactions involved in stimulation of the M phi by endotoxin will lead to therapies to modulate the response and prevent deleterious processes such as ARDS. In the present studies, LPS from E. coli 0111:B4 was shown in a dose response to stimulate large levels of both PCA and TNF in alveolar M phi. LPS from Bacteroides fragilis and Lipid X (the monosaccharide precursor of endotoxin) were unable to cause stimulation of the M phi in vitro. However, both moieties, B. fragilis LPS and Lipid X, were able to effectively and specifically compete with E. coli LPS and block M phi stimulation.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Endotoxinas/farmacología , Macrófagos/efectos de los fármacos , Insuficiencia Multiorgánica/etiología , Alveolos Pulmonares/efectos de los fármacos , Animales , Bacteroides fragilis , Factores de Coagulación Sanguínea/biosíntesis , Células Cultivadas , Escherichia coli , Glucolípidos/metabolismo , Humanos , Lipopolisacáridos/metabolismo , Activación de Macrófagos , Macrófagos/fisiología , Masculino , Insuficiencia Multiorgánica/fisiopatología , Alveolos Pulmonares/metabolismo , Conejos , Síndrome de Dificultad Respiratoria/etiología , Factor de Necrosis Tumoral alfa/biosíntesis
17.
Infect Immun ; 63(10): 4046-53, 1995 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7558318

RESUMEN

In response to bacterial lipopolysaccharides (LPS; endotoxin), endothelial cells are converted to an activation phenotype expressing both proinflammatory and procoagulant properties that include the induction of leukocyte adhesion molecules and tissue factor expression. LPS-induced endothelial cell activation requires a soluble form of the monocyte LPS receptor, sCD14. We evaluated the capacity of multiple strains of gram-negative and gram-positive bacteria to induce endothelial E-selectin and tissue factor expression through sCD14-dependent pathways with cultured human umbilical vein endothelial cells (HUVE). Both viable and heat-killed gram-negative bacteria (Bacteroides fragilis, Enterobacter cloacae, Haemophilus influenzae, and Klebsiella pneumoniae) but not viable or heat-killed gram-positive bacteria (Staphylococcus aureus, Enterococcus faecalis, and Streptococcus pneumoniae) induced prominent E-selectin surface expression detected by enzyme-linked immunosorbent assay. Tissue factor activity on HUVE, indicated by factor X activation, was induced in response to gram-negative bacteria but not in response to gram-positive bacteria. Gram-negative bacteria induced transcriptional activation in HUVE, indicated by the appearance of E-selectin-specific mRNA and by the demonstration of activation of NF-kappa B, a trans-activating factor necessary for E-selectin and tissue factor gene transcription. In contrast, neither E-selectin mRNA nor activation of NF-kappa B was detected in HUVE treated with gram-positive bacteria. Endothelial cell activation by gram-negative bacteria in each of these assays was inhibited with a monoclonal antibody (60bd) against CD14. Furthermore, CHO-K1 cells, transfected with human recombinant CD14, responded to all strains of gram-negative bacteria (viable or heat killed), indicated by CHO-K1 NF-kappa B activation. We conclude that gram-negative bacteria induce endothelial cell activation through a common sCD14-dependent pathway.


Asunto(s)
Endotelio Vascular/fisiología , Bacterias Gramnegativas/fisiología , Receptores de Lipopolisacáridos/fisiología , Animales , Secuencia de Bases , Células CHO , Cricetinae , Selectina E/biosíntesis , Endotelio Vascular/citología , Calor , Humanos , Lipopolisacáridos/toxicidad , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Tromboplastina/biosíntesis , Activación Transcripcional , Transfección
18.
Biochem Biophys Res Commun ; 193(3): 1283-90, 1993 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-7686753

RESUMEN

A protein kinase C (PKC) agonist selective for the beta I isozyme, 12-deoxyphorbol 13-phenylacetate 20-acetate (dPPA), induced NF-kappa B-like binding activity and surface expression of E-selectin and VCAM-1 in human umbilical vein endothelial cells (HUVEC), similar to the effects of tumor necrosis factor-alpha (TNF-alpha). Induction of E-selectin and VCAM-1 expression by dPPA was completely inhibited by the PKC inhibitors staurosporine and Ro31-7549. The PKC inhibitors also reduce TNF-alpha-induced VCAM-1 expression. However, neither dPPA nor TNF-alpha translocated PKC from the cytosolic to the plasma or nuclear membrane particulate fractions in HUVEC. These results indicate that activation of the beta I PKC isozyme is sufficient for expression of E-selectin and VCAM-1, and suggest that PKC may mediate the effects of TNF-alpha and dPPA without requiring the translocation normally associated with activation of PKC.


Asunto(s)
Moléculas de Adhesión Celular/biosíntesis , Endotelio Vascular/metabolismo , Isoenzimas/metabolismo , Ésteres del Forbol/farmacología , Proteína Quinasa C/metabolismo , Alcaloides/farmacología , Secuencia de Bases , Moléculas de Adhesión Celular/genética , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , Citosol/enzimología , Selectina E , Endotelio Vascular/efectos de los fármacos , Humanos , Indoles/farmacología , Isoenzimas/antagonistas & inhibidores , Cinética , Maleimidas/farmacología , Glicoproteínas de Membrana/biosíntesis , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Sondas de Oligonucleótidos , Regiones Promotoras Genéticas , Proteína Quinasa C/antagonistas & inhibidores , Estaurosporina , Acetato de Tetradecanoilforbol/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Venas Umbilicales , Molécula 1 de Adhesión Celular Vascular
19.
J Immunol ; 143(9): 3025-30, 1989 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-2681420

RESUMEN

Recent studies in vitro have demonstrated that LPS from Gram-negative bacteria are capable of inducing endothelial cells to express a cell surface property that promotes the adherence of neutrophils (polymorphonuclear cells, PMN). We have investigated the effects of LPS from Bacteroides fragilis, an organism documented to have little toxicity in vivo, on the induction of this property in human endothelial cells. Monolayers of cultured human umbilical vein endothelial cells (HUVE) exhibited no increase in adhesiveness for 51Cr-radiolabeled PMN after 4 h of exposure to B. fragilis LPS from 1 ng to 10 micrograms/ml. Escherichia coli LPS elicited a dose-dependent enhancement of HUVE adhesiveness for PMN over the same concentration range, reaching a maximum of 49.4 +/- 6.6% at 10 micrograms/ml. Like E. coli LPS, B. fragilis LPS converted chromogenic substrate in the Limulus amebocyte lysate assay, and was directly cytotoxic to bovine aortic endothelial cells. Both B. fragilis LPS activities required doses two-to-three log-fold higher than for E. coli LPS. In addition, we found that B. fragilis LPS inhibited the induction of HUVE adhesiveness for PMN by E. coli LPS. This inhibition was also dose-dependent, becoming maximal (greater than 80%) when B. fragilis LPS was in 10- to 20-fold excess. Tumor necrosis factor and IL-1, two monokines which also elicit HUVE adhesiveness for PMN, were not inhibited by B. fragilis LPS, suggesting a mechanism of HUVE activation by LPS which is signal-specific, and which recognizes specificities of LPS structure.


Asunto(s)
Adhesión Celular/efectos de los fármacos , Endotelio Vascular/citología , Lipopolisacáridos/farmacología , Neutrófilos/citología , Bacteroides fragilis , Células Cultivadas , Escherichia coli , Humanos , Interleucina-1/farmacología , Lípido A , Relación Estructura-Actividad , Factor de Necrosis Tumoral alfa/farmacología
20.
J Immunol ; 136(12): 4548-53, 1986 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-3486903

RESUMEN

We examined the role of the neutrophil membrane antigen complex designated CDw18 (LFA-1/Mac-1/p150, 95) in human peripheral blood neutrophil adherence to cultured human umbilical vein endothelial cells (HEC) pretreated with lipopolysaccharide (LPS), interleukin 1 (IL 1), or recombinant tumor necrosis factor-alpha (rTNF-alpha). Pretreatment of HEC with LPS produced a dose-and time-dependent increase in subsequent neutrophil adherence (7 +/- 1% adherence to untreated HEC vs 38 +/- 3% adherence to HEC pretreated for 4 hr with LPS 150 ng/ml; mean +/- SE of 22 experiments: p less than 0.001). This effect was observed in primary and passaged HEC, but not in bovine aortic endothelial cells or human dermal fibroblasts. The LPS-induced activity appeared to be associated with the HEC surface, since it was not removed by washing and was not detected in the supernatant medium. Inhibition of RNA or protein synthesis during pretreatment of HEC with LPS prevented induction of the adherence-promoting activity. Pretreatment of HEC with IL 1 and rTNF-alpha produced a similar protein synthesis-dependent increase in neutrophil adherence to HEC. Coincubation of neutrophils with murine monoclonal antibody (MoAb) 60.3, an antibody directed to the CDw18 complex, produced a 70 +/- 4% inhibition of neutrophil adherence to LPS-pretreated HEC, 59 +/- 5% inhibition of adherence to IL 1-pretreated HEC, and 65 +/- 11% inhibition of adherence to rTNF-alpha-pretreated HEC (means +/- SE of 18, seven, and five experiments, respectively). Notably, MoAb 60.3 did not completely inhibit neutrophil adherence to pretreated HEC, although it completely inhibited adherence to untreated HEC when neutrophils were activated directly with phorbol ester. Similarly, the adherence of neutrophils from a patient with an inherited deficiency of the CDw18 complex to LPS-, IL 1-, and rTNF-alpha-pretreated HEC was markedly reduced compared with normal neutrophils (5 to 11% adherence with CDw18-deficient neutrophils vs 43 to 54% adherence with normal neutrophils), but adherence to pretreated HEC was still significantly greater than adherence to HEC that were not pretreated (2% adherence). We conclude that LPS, IL 1, and rTNF-alpha induce synthesis of an endothelial cell-surface factor(s) that promotes neutrophil adherence primarily by a mechanism involving the CDw18 complex. It thus appears that the CDw18 complex is important for augmented neutrophil adherence to endothelium in vitro whether the is stimulated directly by inflammatory mediators or indirectly by endothelial-dependent mechanisms.


Asunto(s)
Antígenos de Superficie/inmunología , Endotelio/metabolismo , Glicoproteínas/farmacología , Interleucina-1/fisiología , Lipopolisacáridos/farmacología , Neutrófilos/fisiología , Animales , Antígenos de Superficie/biosíntesis , Aorta , Bovinos , Adhesión Celular , Moléculas de Adhesión Celular , Endotelio/inmunología , Granulocitos , Humanos , Isoantígenos/inmunología , Neutrófilos/inmunología , Proteínas Recombinantes/farmacología , Factor de Necrosis Tumoral alfa , Venas Umbilicales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA