Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Trauma ; 50(3): 449-55; discussion 456, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11265023

ABSTRACT

BACKGROUND: Resuscitation with oxygen-carrying fluids is critically important in the patient with hemorrhagic shock caused by trauma. However, it is clear that a number of biologic mediators present in stored blood (packed red blood cells [PRBCs]) have the potential to exacerbate early postinjury hyperinflammation and multiple organ failure through priming of circulating neutrophils (PMNs). PolyHeme (Northfield Laboratories, Evanston, IL), a hemoglobin-based substitute that is free of priming agents, provides an alternative. We hypothesized that PMN priming would be attenuated in patients resuscitated with PolyHeme in lieu of stored blood. METHODS: Injured patients requiring urgent transfusion were given either PolyHeme (up to 20 units) or PRBCs. Early postinjury PMN priming was measured via beta-2 integrin expression, superoxide production, and elastase release. RESULTS: Treatment groups were comparable with respect to extent of injury and early physiologic compromise. PMNs from patients resuscitated with PRBCs showed priming in the early postinjury period by all three measures. No such priming was evident in patients resuscitated with PolyHeme. CONCLUSION: The use of a blood substitute in the early postinjury period avoids PMN priming and may thereby provide an avenue to decrease the incidence or severity of postinjury multiple organ failure.


Subject(s)
Blood Substitutes/therapeutic use , Cytotoxicity, Immunologic/immunology , Hemoglobins/immunology , Hemoglobins/therapeutic use , Multiple Trauma/complications , Multiple Trauma/immunology , Neutrophils/immunology , Pyridoxal Phosphate/analogs & derivatives , Pyridoxal Phosphate/immunology , Pyridoxal Phosphate/therapeutic use , Resuscitation/methods , Shock, Hemorrhagic/etiology , Shock, Hemorrhagic/therapy , Blood Pressure , CD18 Antigens/blood , Humans , Inflammation , Injury Severity Score , Multiple Organ Failure/etiology , Multiple Trauma/classification , Pancreatic Elastase/blood , Prospective Studies , Shock, Hemorrhagic/metabolism , Superoxides/blood , Treatment Outcome
2.
Arch Surg ; 135(2): 219-25, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10668885

ABSTRACT

HYPOTHESIS: Neutrophil priming has been implicated in the development of multiple organ failure, although the precise intracellular mechanisms that regulate neutrophil priming remain unclear. Our previous work characterized platelet-activating factor (PAF) priming of human neutrophils for concordant superoxide anion (O2-) generation and elastase degranulation. The p38 mitogen-activated protein kinase (MAPK) is activated by PAF stimulation. We hypothesized that PAF-induced human neutrophil priming for O2- and elastase release is mediated via the p38 MAPK pathway. DESIGN: Isolated neutrophils from 6 human donors were preincubated with the specific p38 MAPK inhibitor SB 203580 (1 micromol/L) or buffer (control) for 30 minutes. Cells were then primed with PAF (200 nmol/L), followed by receptor-dependent (N-formyl-methionyl-leucyl-phenylalanine, 1 micromol/L) or receptor-independent phorbol myristate acetate (PMA, 100 ng/mL) activation. SETTING: Urban trauma research laboratory. PATIENTS: Healthy volunteer donors of neutrophils. MAIN OUTCOME MEASURES: Maximal rate of O2- generation was measured by superoxide dismutase-inhibitable reduction of cytochrome c and elastase release by the cleavage of N-methoxysuccinyl-Ala-Ala-Pro-Val-p-nitroanilide. RESULTS: SB 203580 significantly attenuated the generation of O2- and release of elastase from neutrophils activated with N-formyl-methionyl-leucyl-phenylalanine but not with PMA. Independent of the activator receptor status, SB 203580 almost completely blocked the exaggerated neutrophil cytotoxic response due to PAF priming. CONCLUSIONS: The p38 MAPK pathway is required for maximal PAF-induced neutrophil priming for O2- production and elastase degranulation. Therefore, the MAPK signaling cascade may offer a potential therapeutic strategy to preempt global neutrophil hyperactivity rather than attempt to nullify the end products independently.


Subject(s)
Leukocyte Elastase/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neutrophil Activation/physiology , Superoxides/metabolism , Enzyme Activation , Humans , Platelet Activating Factor/pharmacology
3.
J Trauma ; 48(1): 45-8, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10647564

ABSTRACT

OBJECTIVE: Neutrophil (PMN) priming after hemorrhagic shock is predictive of the subsequent development of multiple organ failure, but the mechanism remains unknown. Recently, we and others have demonstrated that mesenteric lymph from shock animals resuscitated with lactated Ringer's solution (LR) is not only a potent PMN priming agent but also causes lung injury. Work by others has shown that resuscitation with hypertonic saline (HTS) protects animals from lung injury after hemorrhagic shock. Therefore, we hypothesize that resuscitation with HTS will abolish PMN priming by postshock mesenteric lymph. METHODS: After mesenteric lymph duct catheterization, male rats underwent hemorrhagic shock (mean arterial pressure of 40 mm Hg for 90 minutes) and resuscitation with shed blood plus either LR (2x volume of shed blood) or 4 mL/kg of 7% HTS (isonatremic). Priming for superoxide by PMN was measured after fMLP (1 microM) activation. RESULTS: Shock significantly decreased mesenteric lymph flow from preshock levels in both groups. LR resuscitation produced significantly more mesenteric lymph than HTS resuscitation. Mesenteric lymph from LR animals primed PMN for superoxide production, whereas, HTS eliminated this priming. CONCLUSION: HTS not only decreases postshock mesenteric lymph production, it eliminates PMN priming by mesenteric lymph, suggesting a mechanism for the beneficial effects of HTS resuscitation.


Subject(s)
Lymph/immunology , Mesentery , Neutrophils/immunology , Resuscitation/methods , Saline Solution, Hypertonic/therapeutic use , Shock, Hemorrhagic/immunology , Shock, Hemorrhagic/therapy , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Isotonic Solutions/therapeutic use , Lymph/chemistry , Male , Multiple Organ Failure/etiology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Predictive Value of Tests , Rats , Rats, Sprague-Dawley , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/prevention & control , Ringer's Lactate , Shock, Hemorrhagic/complications , Shock, Hemorrhagic/physiopathology , Splanchnic Circulation/drug effects , Superoxides/analysis
4.
Arch Surg ; 134(10): 1074-8, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10522849

ABSTRACT

BACKGROUND: Inflammatory stimuli rapidly activate mitogen-activated protein kinases (MAPKs) in neutrophils (PMNs). However, their role in cytotoxic function remains unknown. Elucidating the signals involved in release of cytotoxic agents from PMNs may provide new avenues for therapy in diseases of diminished or excessive PMN function. HYPOTHESIS: The p38 MAPK and extracellular signal-related kinase 1/2 (ERK1/2) modulate superoxide generation and elastase release in activated human PMNs. STUDY DESIGN: Isolated human PMNs were incubated with specific inhibitors of MAPK pathways, or vehicle control solution, before activation with the bacterial peptide f-Met-Leu-Phe. MAIN OUTCOME MEASURES: The rate of superoxide release from activated PMNs was measured by the superoxide dismutase-inhibitable reduction of cytochrome-c. Elastase release from PMNs was determined by cleavage of the substrate Ala-Ala-Pro-Val-pNA. RESULTS: Superoxide release from activated PMNs was inhibited by blockade of p38 MAPK activation but unaffected by blockade of ERK1/2. Conversely, elastase release was unaffected by p38 MAPK inhibition and increased by ERK1/2 inhibition. CONCLUSIONS: Activation of p38 MAPK promotes superoxide release from PMNs activated by f-Met-Leu-Phe. The ERK1/2 pathway may serve as a negative feedback mechanism for granule exocytosis.


Subject(s)
Mitogen-Activated Protein Kinases/physiology , Neutrophils/immunology , Cytotoxicity, Immunologic , Humans , Mitogen-Activated Protein Kinase 3 , Pancreatic Elastase/metabolism , Superoxides/metabolism , p38 Mitogen-Activated Protein Kinases
5.
J Surg Res ; 83(2): 83-8, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10329099

ABSTRACT

Mesenteric lymph has recently been invoked as an avenue for gut-derived factors that may result in distant organ injury following hemorrhagic shock. We demonstrate that posthemorrhagic shock mesenteric lymph primes neutrophils (PMNs) and causes lung injury. Methods. Mesenteric lymph was collected from Sprague-Dawley rats from their mesenteric lymph duct prior to, during, and following hemorrhagic shock (MAP 40 for 90 min). The rats were then resuscitated with shed blood plus lactated Ringers (2X shed blood) over 3 h. Lung leak was assessed by transudation of Evan's blue dye into the alveolus as measured by bronchoalveolar lavage. Isolated human PMNs were incubated with 1 and 10% lymph; priming was measured by the fMLP (1 microM)-stimulated production of superoxide and surface expression of CD11b determined by flow cytometry. Results. Mesenteric lymph flow increased significantly during resuscitation: preshock 144.4 microl/h, shock 44.5 microl/h, resuscitation 566.6 microl/h. Furthermore, diversion of this lymph abrogated lung injury as compared to rats without lymph diversion. Finally, mesenteric lymph from postshock animals primed PMNs for superoxide production (nearly three times control cells) as well as increased surface expression of CD11b (2-fold over control). Conclusion. Mesenteric lymph primes PMNs and causes lung injury following hemorrhagic shock. Mesenteric lymph provides a conduit for proinflammatory mediators that may participate in the pathogenesis of MOF.


Subject(s)
Lung/physiopathology , Lymph/physiology , Neutrophils/physiology , Shock, Hemorrhagic/physiopathology , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Flow Cytometry , Gene Expression Regulation , Humans , Lung Injury , Macrophage-1 Antigen/blood , Macrophage-1 Antigen/genetics , Male , Mesentery , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Rats , Rats, Sprague-Dawley , Superoxides/blood
6.
J Leukoc Biol ; 65(2): 211-6, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10088604

ABSTRACT

The importance of reactive oxygen species (ROS) in neutrophil (PMN)-mediated injury to host tissues has been strongly implicated in a number of animal models. Peculiarities of the laboratory rat PMN, including an apparent paucity of superoxide release, prompted us to examine disparities in the respiratory burst between human and rat PMNs. Using isolated PMNs, we examined oxygen consumption, superoxide release, nitrate/nitrite release, and dihydrorhodamine (DHR) oxidation in response to an array of soluble stimuli. Our findings confirm that intact rat PMNs release little superoxide in comparison to human PMNs when primed and activated by soluble stimuli. For example, PMA-activated human PMNs released superoxide at 10.1 +/- 2.7 times the rate of rat PMNs (P < 0.01). However, measurements of oxygen consumption, cell-associated oxidant production (by DHR oxidation) and release of superoxide from electroporated cells suggests that rat PMNs generate oxidants at rates equivalent to human PMNs but preferentially release them in an intracellular compartment. Implications for the study of PMN-mediated oxidant injury in animal models are discussed.


Subject(s)
Neutrophils/physiology , Respiratory Burst/physiology , Animals , Humans , Nitrates/metabolism , Nitrites/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Rhodamines/metabolism , Species Specificity , Superoxides/metabolism
7.
J Trauma ; 46(1): 42-8, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9932682

ABSTRACT

BACKGROUND: Postinjury neutrophil (PMN) priming identifies the injured patient at risk for the subsequent development of multiple organ failure (MOF). PMN priming has previously been shown to cause enhanced release of proteases and superoxide. PMNs, however, are a rich source of proinflammatory cytokines, such as interleukin (IL)-8 and tumor necrosis factor (TNF), which have been implicated in the development of MOF. PMNs also make IL-1ra, which is an anti-inflammatory cytokine that inhibits IL-1. It is our hypothesis that postinjury PMNs are primed for increased stimulated release of the proinflammatory cytokines IL-8 and TNF but not the anti-inflammatory cytokine IL-1ra. METHODS: Twelve trauma patients with a mean Injury Severity Score of 24 (+/-4.6) and 10 elective surgical patients were studied. Postinjury PMNs were isolated from blood obtained at presentation (within 2 hours after injury) and 24 hours after trauma. PMNs from elective surgical patients were obtained preoperatively, immediately postoperatively, and at 24 hours. The PMNs were stimulated with platelet-activating factor (200 nM)/N-formyl-methionyl-leucyl-phenylalanine (1 micromol/L) or lipopolysaccharide (100 ng/mL) incubated for 24 hours in RPMI-1640, and release of IL-8, TNF, and IL-1ra were measured. RESULTS: Postinjury PMNs were primed for both platelet-activating factor/N-formyl-methionyl-leucyl-phenylalanine-stimulated and lipopolysaccharide-stimulated IL-8 and TNF release at 2 hours after injury (fourfold increase of IL-8 release and fivefold increase of TNF release), whereas elective surgical patients demonstrated no priming. In contrast, postinjury patients were not primed for increased release of the counterinflammatory cytokine IL-1ra, suggesting a specific postinjury up-regulation of IL-8 and TNF. CONCLUSION: After injury, PMNs are primed for proinflammatory cytokine release in addition to superoxide and elastase. This augmented release of IL-8 and TNF may be involved in the subsequent development of organ dysfunction and ultimately MOF.


Subject(s)
Cytokines/metabolism , Multiple Organ Failure/immunology , Neutrophils/metabolism , Wounds and Injuries/immunology , Adult , Case-Control Studies , Female , Humans , Injury Severity Score , Interleukin-1/metabolism , Interleukin-8/metabolism , Male , Tumor Necrosis Factor-alpha/metabolism
8.
Shock ; 11(2): 77-81, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10030791

ABSTRACT

UNLABELLED: Postinjury neutrophil (PMN) dysfunction is a well recognized event that may be responsible for increased infections. PMN cytokine production is an important component of their bactericidal capacity. When PMNs are stimulated, inhibitory factor kappaB (IkappaB) is degraded, allowing nuclear factor kappaB (NFkappaB) to translocate to the nucleus and promotes genes for the transcription of the interleukin-8 (IL-8) and tumor necrosis factor (TNF) genes. We hypothesize that similar to their late postinjury depressed superoxide production, postinjury PMNs manifest suppressed cytokine production, which is mediated by stabilization of IkappaB levels. METHODS: Twelve severely injured patients with an injury severity score (ISS) of 24 (+/-4.6) were studied as well as 10 elective surgical patients as a control. PMNs were isolated and incubated for 24 h in RPMI. PMNs were stimulated with lipopolysaccharide (LPS; 100 ng) or PAF (200 nm) and fMLP (1 microM) and release of IL-8, TNF, and interleukin-1 receptor antagonist (IL-1ra) were measured. Postinjury PMNs were also stimulated with LPS (100 ng), and IkappaB breakdown was measured at 0, 30, and 60 min using gel electrophoresis. RESULTS: Postinjury PMNs displayed a significant suppression of both IL-8 and TNF on postinjury Days 1-3, while the release of IL-1ra was preserved throughout the entire study period. In contrast, elective surgical patients demonstrated no decrease in IL-8 or TNF. Furthermore, IkappaB levels were preserved in the postinjury PMNs as compared with normal control PMNs. CONCLUSION: Postinjury PMNs have a suppressed release of both IL-8 and TNF following injury that did not occur in elective surgical patients. Furthermore, the NFkappaB/IkappaB-independent IL-1ra did not show suppression of release. In addition, stabilization of IkappaB following severe injury leads to decreased PMN IL-8 and TNF production. This genetic reprogramming may help explain PMN dysfunction and subsequent infections seen in severely injured patients.


Subject(s)
Cytokines/blood , DNA-Binding Proteins/blood , Neutrophils/metabolism , Wounds, Nonpenetrating/blood , Wounds, Penetrating/blood , Adult , Humans , I-kappa B Proteins , In Vitro Techniques , Interleukin 1 Receptor Antagonist Protein , Interleukin-8/blood , Lipopolysaccharides/pharmacology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Platelet Activating Factor/pharmacology , Postoperative Complications/blood , Sialoglycoproteins/blood , Tumor Necrosis Factor-alpha/metabolism
9.
Arch Surg ; 133(11): 1229-33, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9820355

ABSTRACT

BACKGROUND: Secretory phospholipase A2 (sPLA2) is a potent proinflammatory enzyme that stimulates inflammation through the production of reactive lipids. However, enzymatic inhibitors have been disappointing in their effectiveness in halting hyperinflammation. OBJECTIVE: To determine whether sPLA2 acts directly on neutrophil plasma membrane lipids or via a nonenzymatic mechanism. DESIGN: Isolated neutrophils (PMNs) were incubated with 3 types of sPLA2, and elastase and superoxide release from PMNs was measured. Ethyleneglycotetraacetic acid was used as a selective enzymatic inhibitor. The PMNs were exposed to sPLA2 in the presence and absence of ethyleneglycotetraacetic acid and the release of elastase was measured. SETTING: Urban trauma research laboratory. PATIENTS: Normal healthy donors of PMNs. MAIN OUTCOME MEASURES: Stimulated release of superoxide and elastase. RESULTS: The sPLA2 acted directly on plasma membrane lipids to stimulate the PMN to produce superoxide and release elastase. This mechanism is blocked with enzymatic inhibition of sPLA2. The sPLA2 also provokes elastase release from PMNs independently of its enzymatic function. This mechanism is not blocked with traditional enzymatic inhibitors. CONCLUSIONS: These data indicate that the sPLA2 can act directly on PMNs to stimulate the release of inflammatory mediators via enzymatic degradation of plasma membrane lipids. In addition, sPLA2 can act as a ligand and stimulate the PMN independently of its enzymatic activity.


Subject(s)
Inflammation/immunology , Neutrophil Activation/immunology , Neutrophils/metabolism , Pancreatic Elastase/immunology , Phospholipases A/physiology , Reactive Oxygen Species/immunology , Superoxides/immunology , Animals , Egtazic Acid/pharmacology , Elapidae/immunology , Group II Phospholipases A2 , Humans , Multiple Organ Failure/immunology , Phospholipases A2 , Respiratory Burst/immunology
10.
J Surg Res ; 76(1): 91-4, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9695746

ABSTRACT

BACKGROUND: Interleukin-6 (IL-6) appears to be a reliable marker of disease severity in critically ill patients at risk for inflammatory organ injury such as ARDS and MOF. Debate continues, however, as to whether this pleiotropic cytokine acts principally as a proinflammatory or counterregulatory mediator. Because the polymorphonuclear leukocyte (PMN) is a central effector of inflammatory injury, defining the effects of IL-6 on mechanisms of PMN cytotoxicity may be revealing. Previous investigations of PMN release of reactive oxygen species demonstrate that IL-6 in concert with other mediators may augment cytotoxicity. We hypothesized that IL-6 alone increases PMN cytotoxic potential through selective enhancement of elastase release. MATERIALS AND METHODS: Isolated human PMNs were incubated with IL-6 in the physiologic range observed in critically ill patients (0.1 to 100 ng/ml) for 10 to 30 min. Selected cells were then activated with fMLP (1 microM). Elastase release was measured by specific cleavage of AAPV-pNA and compared to untreated cells and cells activated with formyl-Met-Leu-Phe (fMLP; 1 microM) alone. To determine if changes in elastase release might be due to IL-6 induced generation of PAF, WEB 2347 (50 microM) was preincubated with selected cells for 20 min. Surface expression of beta 2 integrins was measured by flow cytometry after incubating with labeled antibodies to CD11b and CD18. RESULTS: IL-6 alone at 100 ng/ml augmented basal elastase release by 116 +/- 41% within 10 min. Doses as low as 0.1 ng/ml stimulated elastase release when the incubation time was increased to 30 min. After 30 min of incubation, IL-6 at all doses examined augmented the elastase release of fMLP-activated cells (increases of 33 to 45%). WEB 2347 preincubation did not block augmentation of elastase release by IL-6 at 10 ng/ml. IL-6 had no effect on surface expression of beta 2 integrins at 10 ng/ml. CONCLUSIONS: IL-6 alone enhances both basal and fMLP-stimulated elastase release by PMNs. This proinflammatory action on PMNs may help explain the observed correlation between circulating IL-6 levels and inflammatory organ injury.


Subject(s)
Interleukin-6/pharmacology , Neutrophil Activation/immunology , Neutrophils/enzymology , Pancreatic Elastase/metabolism , Receptors, G-Protein-Coupled , CD18 Antigens/metabolism , Dose-Response Relationship, Immunologic , Humans , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophil Activation/drug effects , Neutrophils/immunology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptors, Cell Surface/metabolism
11.
J Surg Res ; 77(1): 71-4, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9698536

ABSTRACT

BACKGROUND: Priming of neutrophils (PMNs) for protease release is believed to be central to the pathogenesis of PMN-mediated tissue injury observed in ARDS/MOF. Defining the intracellular signaling pathways involved with this excessive protease release may aid in establishing future therapies for ARDS. Phospholipase D (PLD) production of phosphatidic acid (PA) is thought to be pivotal in reactive oxygen species generation but its role in degranulation (i. e., protease release) remains unclear. Our hypothesis was that primed neutrophils require PLD production of PA for maximal activated release of elastase. METHODS: Isolated human PMNs were incubated with a well described antagonist of PA production, ethanol (ETOH, 100-1000 mg/dL), and then primed (PAF, 200 nM) followed by activation (fMLP, 1 microM). To mimic fMLP receptor-dependent activation, PMNs were primed and then directly activated with exogenous dioctanoyl l-alpha-phosphatidic acid (PA8, 0.5-200 microM). To confirm the importance of PA in elastase release, PA8 was given to primed-activated PMNs after ethanol pretreatment in an attempt to recover the maximal response. Elastase release was measured by the cleavage of AAPV-pNA. RESULTS: PA blockade with ETOH attenuated PAF-primed/fMLP-activated PMN elastase release in a dose-dependent manner. Exogenous PA8 reproduced maximally primed-activated PMN elastase release, and furthermore, PA8 was able to restore maximal elastase release following ethanol attenuation. CONCLUSIONS: Elastase release from PAF-primed/ fMLP-activated neutrophils is dependent on PA production. Thus, PA production appears to be involved in both oxidant-dependent and independent mechanisms of neutrophil cytotoxicity and may be a potential therapeutic target in the treatment of hyperinflammatory diseases such as ARDS/MOF.


Subject(s)
Leukocyte Elastase/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Phosphatidic Acids/pharmacology , Receptors, Immunologic/physiology , Receptors, Peptide/physiology , Humans , Phosphatidic Acids/metabolism , Receptors, Formyl Peptide
12.
Surgery ; 124(2): 403-7; discussion 408, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9706165

ABSTRACT

BACKGROUND: Increased expression of pulmonary endothelial intercellular adhesion molecule-1 (ICAM-1) is obligatory to neutrophil adherence culminating in adult respiratory distress syndrome (ARDS). The p38 mitogen-activated protein kinases (MAPKs) have been established as crucial in leukocyte proinflammatory signaling, but their role in the endothelial cells remains ill defined. We hypothesized that p38 MAPK activity is integral to ICAM-1 up-regulation on pulmonary endothelium. METHODS: Human pulmonary microvascular endothelial cells (HMVECs) were grown to confluence and pretreated with either the tyrosine phosphorylation inhibitor herbimycin A (1 mumol/L or the p38 MAPK inhibitor SB 203580 (10(-7) to 10(-5) mol /L) for 6 hours. ICAM-1 expression was quantified by flow cytometry. Data expressed as mean fluorescence intensity. Western blotting was used to show p38 MAPK activity after stimulation with lipopolysaccharide (LOS) or tumor necrosis factor-alpha (TNF-alpha). RESULTS: Tyrosine phosphorylation inhibition with herbimycin A attenuated both LPS and TNF-alpha stimulated ICAM-1 up-regulation. Similarly, specific inhibition of p38 MAPK attenuated both LPS (10(-6) to 10(-5) mol/L SB203580) and TNF-alpha (10(-7) to 10(-5) mol/L SB203580) stimulated expression of ICAM-1 on HMVECs. Both LPS and TNF-alpha induced activation of p38 in HMVECs. CONCLUSIONS: Signaling through p 38 MAPKs contributes to LP and TNF-alpha stimulated ICAM-1 surface expression on HMVECs. Thus p38 MAPKs appear integral to both neutrophil and endothelial cell proinflammatory signaling and may be a potential therapeutic target in the treatment of ARDS.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Endothelium, Vascular/enzymology , Intercellular Adhesion Molecule-1/metabolism , Mitogen-Activated Protein Kinases , Up-Regulation/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Capillaries/chemistry , Capillaries/cytology , Capillaries/enzymology , Cells, Cultured , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/pharmacology , Lipopolysaccharides/pharmacology , Lung/blood supply , Phosphorylation , Pyridines/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Tyrosine/metabolism , Up-Regulation/physiology , p38 Mitogen-Activated Protein Kinases
13.
J Trauma ; 44(4): 592-7; discussion 598, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9555828

ABSTRACT

BACKGROUND: Ongoing clinical trials have revived interest in hypertonic saline (HTS) for postinjury resuscitation; these studies have documented serum Na+ concentrations > or = 170 mmol/L. Recent animal studies have shown that HTS enhances T-cell and monocyte function, but effects on the polymorphonuclear neutrophil (PMN) remain unclear. The postinjury lipid mediators platelet-activating factor (PAF) and leukotriene B4 (LTB4) have been implicated in PMN priming for cytotoxicity, which is believed to be important in the pathogenesis of multiple organ failure. We hypothesized that HTS would stimulate PMN superoxide (O2-) and elastase release from PAF- and LTB4-primed PMNs. METHODS: Isolated PMNs from five donors were primed for 5 minutes with 200 nmol/L PAF or 1 micromol/L LTB4 in Kreb's-Ringer's phosphate with dextrose at a Na+ concentration of 140 mmol/L (normal serum Na+ concentration), pelleted, and resuspended in Kreb's-Ringer's phosphate with dextrose for 10 minutes at a Na+ concentration of 130 to 170 mmol/L. O2- generation was measured by superoxide dismutase-inhibitable reduction of cytochrome c and elastase release by cleavage of N-methoxysuccinyl-Ala-Ala-Pro-Val p-nitroanilide. RESULTS: HTS with Na+ concentration up to 170 mmol/L had no significant effect on O2- production or elastase release from quiescent cells. Na+ concentration of 160 and 170 mmol/L, however, activated PAF- and LTB4-primed PMNs for enhanced elastase release with no effect on O2- production. CONCLUSION: In clinically relevant concentrations, elevated Na+ activates lipid-primed neutrophils for enhanced elastase degranulation. Consequently, the administration of HTS in the early postinjury resuscitation period, when PMNs are maximally primed, may activate PMN elastase release and thereby promote the development of multiple organ failure.


Subject(s)
Leukotriene B4/physiology , Neutrophil Activation/drug effects , Neutrophils/drug effects , Pancreatic Elastase/metabolism , Platelet Activating Factor/physiology , Saline Solution, Hypertonic/therapeutic use , Humans , Multiple Organ Failure/etiology , Multiple Trauma/complications , Multiple Trauma/immunology , Multiple Trauma/therapy , Neutrophils/metabolism , Osmolar Concentration , Resuscitation , Superoxides/metabolism
14.
Shock ; 9(3): 171-6, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9525323

ABSTRACT

In a recent clinical trial, liposomal prostaglandin E1 (PGE1) improved oxygenation, increased compliance, and decreased ventilator dependency in patients with adult respiratory distress syndrome (ARDS), thus renewing interest in PGE1 as a potential modulator of inflammation. The neutrophil (PMN) is believed to play a key role in the development of ARDS. Consequently, we investigated the effects of PGE1 on three components of the neutrophil inflammatory response: reactive oxygen species (ROS) generation, protease release, and surface expression of adhesion molecules. Human neutrophils were incubated with PGE1 and then primed with platelet-activating factor (PAF) and activation with N-formyl-methionyl-leucylphenylalanine (fMLP). PGE1 at a dose range of (10[-8] to 10[-5] M) attenuated primed/activated (PAF/fMLP) PMN superoxide anion generation and elastase release. In contrast, PGE1 doses > or =10[-5] M were required to attenuate PAF-stimulated neutrophil upregulation of CD11b/CD18 adhesion molecules. PGE1 also diminished the duration of the PAF-induced cytosolic calcium (Ca2+) flux. Our results suggest that plasma levels of PGE1 attained in patients with ARDS may attenuate ROS and protease neutrophil cytotoxicity but may not effectively block PMN-endothelial cell (EC) adhesion. This attenuation may occur through abrogation of the Ca2+ influx.


Subject(s)
Alprostadil/pharmacology , Macrophage-1 Antigen/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Pancreatic Elastase/metabolism , CD18 Antigens/drug effects , CD18 Antigens/metabolism , Calcium/metabolism , Humans , Macrophage-1 Antigen/drug effects , Superoxides/metabolism , Up-Regulation
15.
J Trauma ; 44(2): 320-4, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9498504

ABSTRACT

BACKGROUND: Acute alcohol intoxication is associated with an increased risk of infection in the injured patient. The impact of clinically relevant levels of ethanol (ETOH) on neutrophil (PMN) bactericidal activity remains ill-defined. PMN priming optimizes microbicidal activity by enhancing oxygen radical production, degranulation, and adhesion molecule up-regulation. We hypothesized that clinically relevant levels of ETOH attenuate these primed PMN responses critical to eradicate infection. METHODS: After incubation with ETOH (0-1.0%), isolated human PMNs were primed with beta-acetyl-gamma-O-alkyl and activated with N-formyl-methionyl-leucyl-phenylalanine. Superoxide generation was measured by cytochrome c reduction, elastase release was measured by cleavage of methoxysuccinyl-ala-ala-pro-val-p-nitroanilide, and CD11b was measured by fluorescent monoclonal antibody staining. Bactericidal activity was assessed by Staphylococcus aureus killing. RESULTS: ETOH attenuated superoxide production dose-dependently with significance at 0.3% ETOH. Elastase release was attenuated starting at 0.2% ETOH, and CD11b expression was reduced starting at 0.4% ETOH. S. aureus killing was impaired dose-dependently with significance at 0.3% ETOH. CONCLUSION: Clinically relevant concentrations of ETOH attenuate PMN functions critical in host defense against invading pathogens. These results provide direct in vitro evidence consistent with previous in vivo data that acute alcohol intoxication is important in the pathogenesis of trauma-related infections.


Subject(s)
Blood Bactericidal Activity/drug effects , Ethanol/pharmacology , Neutrophils/drug effects , Alcoholic Intoxication/immunology , Cells, Cultured , Ethanol/immunology , Humans , Leukocyte Elastase/biosynthesis , Macrophage-1 Antigen/metabolism , Neutrophils/enzymology , Neutrophils/metabolism , Superoxides/metabolism , Up-Regulation
16.
Am J Surg ; 176(6): 612-7, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9926800

ABSTRACT

BACKGROUND: Hemoglobin-based blood substitutes appear poised to deliver the promise of a universally compatible, disease-free alternative to banked blood. However, vasoconstriction following administration of tetrameric hemoglobins has been problematic, likely because of nitric oxide binding. Polymerized hemoglobin is effectively excluded from the abluminal space because of its size, and is thus less likely to perturb vasorelaxation. We therefore hypothesized that hemodynamic responses would be no different in injured patients receiving polymerized hemoglobin versus banked blood. METHODS: Injured patients requiring urgent transfusion were randomized to receive either polymerized hemoglobin or banked blood. Systemic arterial pressure, pulmonary arterial pressure, cardiac index, pulmonary capillary wedge pressure, systemic vascular resistance, and pulmonary vascular resistance were measured serially. RESULTS: There was no difference in any of the measured hemodynamic parameters between patients resuscitated with polymerized hemoglobin versus blood. CONCLUSIONS: Polymerized hemoglobin given in large doses to injured patients lacks the vasoconstrictive effects reported in the use of other hemoglobin-based blood substitutes. This supports the continued investigation of polymerized hemoglobin in injured patients requiring urgent transfusion.


Subject(s)
Blood Substitutes , Hemoglobins , Hypertension, Pulmonary/etiology , Polymers , Wounds and Injuries/therapy , Adult , Female , Hemodynamics/drug effects , Humans , Male , Resuscitation/methods , Shock, Hemorrhagic/therapy , Vasoconstriction
17.
Can J Cardiol ; 8(9): 954-60, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1486546

ABSTRACT

OBJECTIVE: To investigate vascular responses to the endothelin-1 (ET-1) precursor, human big endothelin 1-38 (big ET), in the peripheral vascular bed of the cat. DESIGN: These studies were designed to investigate the hypothesis that bit ET is converted to an active peptide with properties similar to ET-1. SETTING: Hindquarters vascular bed of the cat under conditions of controlled bloodflow; changes in perfusion pressure reflect changes in vascular resistance. ANIMALS: Fifty-four adult mongrel cats. INTERVENTIONS: Big ET, ET-1, the peptidases chymotrypsin, pepsin and cathepsin-D, and the metalloprotease inhibitor phosphoramidon. MAIN RESULTS: Intra-arterial injections of big ET induced a slow-developing and sustained increase in hindquarters perfusion pressure which could be blocked by phosphoramidon. ET-1 (0.3 nmol), administered as a slow infusion over a 10-min period, produced a slowly developing increase in hindquarters perfusion pressure in a manner similar to that observed in response to injection of big ET. A bolus injection of ET-1 produced a biphasic response characterized by a transient decrease in pressure followed by an increase which was significantly greater in magnitude and more rapid in onset than the pressor response to big ET (0.3 nmol). After incubation of big ET with chymotrypsin, pepsin and cathepsin-D (each 5% weight/weight) for 30 mins at 37 degrees C, injection of activated big ET produced a biphasic response characteristic of the response to ET-1 with an initial transient decrease in pressure followed by a secondary increase in hindquarters perfusion pressure. CONCLUSIONS: Big ET produces a phosphoramidon-sensitive pressor response which is similar to that produced by an infusion of ET-1. These data suggest that chymotrypsin, pepsin and cathepsin-D can convert big ET to an active peptide which elicits a biphasic response similar to that produced by ET-1.


Subject(s)
Blood Vessels/drug effects , Endothelins/pharmacology , Protein Precursors/pharmacology , Animals , Blood Pressure/drug effects , Blood Vessels/physiology , Cathepsin D/pharmacology , Cats , Chymotrypsin/pharmacology , Dose-Response Relationship, Drug , Drug Combinations , Endothelin-1 , Glycopeptides/pharmacology , Hindlimb/blood supply , Infusions, Intra-Arterial , Injections, Intra-Arterial , Neprilysin/antagonists & inhibitors , Pepsin A/pharmacology , Regional Blood Flow/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...