Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 186
Filter
1.
Anaesthesist ; 66(7): 479-490, 2017 Jul.
Article in German | MEDLINE | ID: mdl-28677016

ABSTRACT

An impairment of cardiac function is a key feature of cardiovascular failure associated with sepsis; however, its clinical relevance is still underestimated. Recent advancements in echocardiography in patients with septic shock enable a better characterization of septic cardiomyopathy by unmasking a severe, cardiac dysfunction even in the presence of preserved left ventricular ejection fraction. The pathophysiology of septic cardiomyopathy involves a complex mixture of systemic factors and molecular, metabolic, and structural changes of the cardiomyocytes. A better understanding of these factors will enable the discovery of new therapeutic targets for urgently needed disease-modifying therapeutic interventions. To date, the cornerstone of therapeutic management lies in control of the underlying infectious process and hemodynamic stabilization. This review summarizes the pathogenesis, diagnosis, and treatment of septic cardiomyopathy, and highlights the importance of further urgently needed studies aimed at improving diagnosis and treatment for septic cardiomyopathy.


Subject(s)
Cardiomyopathies/physiopathology , Heart/physiopathology , Sepsis/physiopathology , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/metabolism , Cardiomyopathies/therapy , Echocardiography , Heart/diagnostic imaging , Humans , Sepsis/diagnostic imaging , Sepsis/metabolism , Sepsis/therapy , Shock, Septic/metabolism , Shock, Septic/physiopathology , Shock, Septic/therapy
2.
Br J Pharmacol ; 171(24): 5802-15, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25164531

ABSTRACT

BACKGROUND AND PURPOSE: The haematopoietic activity of erythropoietin (EPO) is mediated by the classic EPO receptor (EpoR) homodimer, whereas tissue-protective effects are mediated by a heterocomplex between EpoR and the ß-common receptor (ßcR). Here, we investigated the effects of a novel, selective ligand of this heterocomplex - pyroglutamate helix B surface peptide (pHBSP) - in mice fed a diet enriched in sugars and saturated fats. EXPERIMENTAL APPROACH: Male C57BL/6J mice were fed a high-fat high-sucrose diet (HFHS) for 22 weeks. pHBSP (30 µg·kg(-1) s.c.) was administered for the last 11 weeks. Biochemical assays, histopathological and immunohistochemical examinations and Western blotting were performed on serum and target organs (liver, kidney and skeletal muscle). KEY RESULTS: Mice fed with HFHS diet exhibited insulin resistance, hyperlipidaemia, hepatic lipid accumulation and kidney dysfunction. In gastrocnemius muscle, HFHS impaired the insulin signalling pathway and reduced membrane translocation of glucose transporter type 4 and glycogen content. Treatment with pHBSP ameliorated renal function, reduced hepatic lipid deposition, and normalized serum glucose and lipid profiles. These effects were associated with an improvement in insulin sensitivity and glucose uptake in skeletal muscle. Diet-induced overproduction of the myokines IL-6 and fibroblast growth factor-21 were attenuated by pHBSP and, most importantly, pHBSP markedly enhanced mitochondrial biogenesis in skeletal muscle. CONCLUSIONS AND IMPLICATIONS: Chronic treatment of mice with an EPO derivative, devoid of haematopoietic effects, improved metabolic abnormalities induced by a high-fat high-sucrose diet, by affecting several levels of the insulin signalling and inflammatory cascades within skeletal muscle, while enhancing mitochondrial biogenesis.


Subject(s)
Blood Glucose/drug effects , Fatty Liver/metabolism , Hyperlipidemias/metabolism , Insulin Resistance , Kidney/drug effects , Liver/drug effects , Muscle, Skeletal/drug effects , Oligopeptides/pharmacology , Renal Insufficiency/metabolism , Animals , Blood Glucose/metabolism , Dietary Fats/adverse effects , Dietary Sucrose/adverse effects , Erythropoietin/analogs & derivatives , Fatty Liver/chemically induced , Hyperlipidemias/chemically induced , Kidney/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Renal Insufficiency/chemically induced
3.
Br J Surg ; 99 Suppl 1: 12-20, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22441851

ABSTRACT

BACKGROUND: A systemic inflammatory response syndrome (SIRS) is frequently observed after traumatic injury. The response is sterile and the activating stimulus is tissue damage. Endogenous molecules, called alarmins, are reputed to be released by injured tissues but the precise identity of these mediators is unclear. This review summarizes current preclinical and clinical evidence for trauma alarmins and their role in innate immune activation. METHODS: A comprehensive literature review of putative alarmins in tissue damage after traumatic injury was conducted. RESULTS: The presence of SIRS at admission is an independent predictor of mortality after trauma. The primary initiators of the human immune response are unclear. Several endogenous substances display alarmin characteristics in vitro. Preclinical studies demonstrate that blockade of certain endogenous substances can reduce adverse clinical sequelae after traumatic injury. Human evidence for trauma alarmins is extremely limited. CONCLUSION: The magnitude of acute inflammation is predictive of outcome after trauma, suggesting that an early opportunity for immune modulation may exist. An understanding of the mechanisms of innate immune activation following trauma may lead to new therapeutic agents and improved patient survival.


Subject(s)
Immunity, Innate/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Systemic Inflammatory Response Syndrome/immunology , Wounds and Injuries/immunology , Biomarkers/metabolism , Humans , Necrosis/immunology , Stress, Physiological/immunology , Up-Regulation
4.
J Thromb Haemost ; 8(9): 1919-25, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20553376

ABSTRACT

BACKGROUND: Acute traumatic coagulopathy (ATC) is an impairment of hemostasis that occurs early after injury and is associated with a 4-fold higher mortality, increased transfusion requirements and organ failure. OBJECTIVES: The purpose of the present study was to develop a clinically relevant definition of ATC and understand the etiology of this endogenous coagulopathy. PATIENTS/METHODS: We conducted a retrospective cohort study of trauma patients admitted to five international trauma centers and corroborated our findings in a novel rat model of ATC. Coagulation status on emergency department arrival was correlated with trauma and shock severity, mortality and transfusion requirements. 3646 complete records were available for analysis. RESULTS: Patients arriving with a prothrombin time ratio (PTr) > 1.2 had significantly higher mortality and transfusion requirements than patients with a normal PTr (mortality: 22.7% vs. 7.0%; P < 0.001. Packed red blood cells: 3.5 vs. 1.2 units; P < 0.001. Fresh frozen plasma: 2.1 vs. 0.8 units; P < 0.001). The severity of ATC correlated strongly with the combined degree of injury and shock. The rat model controlled for exogenously induced coagulopathy and mirrored the clinical findings. Significant coagulopathy developed only in animals subjected to both trauma and hemorrhagic shock (PTr: 1.30. APTTr: 1.36; both P < 0.001 compared with sham controls). CONCLUSIONS: ATC develops endogenously in response to a combination of tissue damage and shock. It is associated with increased mortality and transfusion requirements in a dose-dependent manner. When defined by standard clotting times, a PTr > 1.2 should be adopted as a clinically relevant definition of ATC.


Subject(s)
Blood Coagulation Disorders/etiology , Wounds and Injuries/complications , Adult , Animals , Blood Coagulation , Blood Transfusion/methods , Cohort Studies , Disease Models, Animal , Female , Humans , Male , Middle Aged , Rats , Retrospective Studies , Shock, Hemorrhagic/diagnosis
5.
Int J Immunopathol Pharmacol ; 23(4): 1033-46, 2010.
Article in English | MEDLINE | ID: mdl-21244753

ABSTRACT

Peroxisome Proliferator-Activated Receptor ß/δ belongs to a family of ligand-activated transcription factors. Recent data have clarified its metabolic roles and enhanced the potential role of this receptor as a pharmacological target. Moreover, although its role in acute inflammation remains unclear, being the nuclear receptor PPAR ß/δ widely expressed in many tissues, including the vascular endothelium, we assume that the infiltration of PMNs into tissues, a prominent feature in inflammation, may also be related to PPAR ß/δ. Mice subjected to intratracheal instillation of bleomycin (BLEO, 1 mg/kg), a glycopeptide produced by the bacterium Streptomyces verticillus, develop lung inflammation and injury characterized by a significant neutrophil infiltration and tissue oedema. Therefore, the aim of this study is to investigate the effects of GW0742, a synthetic high affinity PPAR ß/δ agonist, and its possible role in preventing the advance of inflammatory and apoptotic processes induced by bleomycin, that long-term leads to the appearance of pulmonary fibrosis. Our data showed that GW0742-treatment (0.3 mg/Kg, 10 percent DMSO, i.p.) has therapeutic effects on pulmonary damage, decreasing many inflammatory and apoptotic parameters detected by measurement of: 1) cytokine production; 2) leukocyte accumulation, indirectly measured as decrease of myeloperoxidase (MPO) activity; 3) IkBα degradation and NF-kB nuclear translocation; 4) ERK phosphorylation; 5) stress oxidative by NO formation due to iNOS expression; 6) nitrotyrosine and PAR localization; 7) the degree of apoptosis, evaluated by Bax and Bcl-2 balance, FAS ligand expression and TUNEL staining. Taken together, our results clearly show that GW0742 reduces the lung injury and inflammation due to the intratracheal BLEO--instillation in mice.


Subject(s)
PPAR delta/agonists , PPAR-beta/agonists , Pneumonia/drug therapy , Thiazoles/therapeutic use , Animals , Apoptosis/drug effects , Bleomycin/toxicity , Interleukin-1beta/biosynthesis , MAP Kinase Signaling System/drug effects , Male , Mice , Nitric Oxide/biosynthesis , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
6.
Nitric Oxide ; 22(2): 141-8, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19892029

ABSTRACT

Nitrite is protective against renal ischemia/reperfusion injury (IRI); an effect due to its reduction to nitric oxide (NO). In addition to other reductase pathways, endothelial NO synthase (eNOS) may also facilitate nitrite reduction in ischemic environments. We investigated the role of eNOS in sodium nitrite (60 microM, 10 ml/kg applied topically 1 min before reperfusion)-induced protection against renal IRI in C57/BL6 wild-type (WT) and eNOS knockout (eNOS KO) mice subjected to bilateral renal ischemia (30 min) and reperfusion (24h). Markers of renal dysfunction (plasma [creatinine] and [urea]), damage (tubular histology) and inflammation (cell recruitment) were elevated following IRI in WT mice; effects significantly reduced following nitrite treatment. Chemiluminescence analysis of cortical and medullary sections of the kidney demonstrated rapid (within 1 min) distribution of nitrite following application. Whilst IRI caused a significant (albeit substantially reduced compared to WT mice) elevation of markers of renal dysfunction and damage in eNOS KO mice, the beneficial effects of nitrite were absent or reduced, respectively. Moreover, nitrite treatment enhanced renal dysfunction in the form of increased plasma [creatinine] in eNOS KO mice. Confirmation of nitrite reductase activity of eNOS was provided by demonstration of nitrite (100 microM)-derived NO production by kidney homogenates of WT mice, that was significantly reduced by L-NMMA. L-NMMA was without effect using kidney homogenates of eNOS KO mice. These results support a role for eNOS in the pathways activated during renal IRI and also identify eNOS as a nitrite reductase in ischemic conditions; activity which in part underlies the protective effects of nitrite.


Subject(s)
Kidney Diseases/prevention & control , Nitric Oxide Synthase Type III/metabolism , Nitrites/therapeutic use , Reperfusion Injury/prevention & control , Animals , Kidney Diseases/drug therapy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/deficiency , Nitrites/metabolism , Nitrites/pharmacology , Reperfusion Injury/drug therapy
7.
Int J Immunopathol Pharmacol ; 20(3): 619-30, 2007.
Article in English | MEDLINE | ID: mdl-17880775

ABSTRACT

Glycogen synthase kinase-3 (GSK-3) is an ubiquitous serine-threonine protein kinase that participates in a multitude of cellular processes and has recently been implicated in the pathophysiology of a number of diseases. The aim of this study is to investigate the effects of TDZD-8, a potent and selective GSK-3beta inhibitor, on the development of lung injury caused by administration of bleomycin (BLM). Mice subjected to intra-tracheal administration of BLM developed significant lung injury characterized by marked neutrophil infiltration and tissue edema. An increase in immunoreactivity to nitrotyrosine, iNOS, TNF-alpha and IL-1beta was also observed in the lungs of BLM-treated mice. In contrast, administration of BLM-treated mice with TDZD-8 (1 mg/kg daily) significantly reduced (I) the degree of lung injury, (II) the increase in staining (immunohistochemistry) for myeloperoxidase (MPO), nitrotyrosine, iNOS, TNF-alpha and IL-1beta and (III) the degree of apoptosis, as evaluated by Bax and Bcl-2 immunoreactivity and TUNEL staining. Taken together, these results clearly demonstrate treatment with the GSK-3beta inhibitor TDZD-8 reduces the development of lung injury and inflammation induced by BLM in mice.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Bleomycin/adverse effects , Enzyme Inhibitors/therapeutic use , Glycogen Synthase Kinase 3/antagonists & inhibitors , Lung Diseases/prevention & control , Lung , Thiadiazoles/therapeutic use , Animals , Cytokines/immunology , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3 beta , Lung/drug effects , Lung/enzymology , Lung/immunology , Lung/pathology , Lung Diseases/chemically induced , Male , Mice , Mice, Inbred Strains , Peroxidase/metabolism , Thiadiazoles/pharmacology
8.
Br J Pharmacol ; 149(6): 687-702, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17016509

ABSTRACT

BACKGROUND AND PURPOSE: Glycogen synthase kinase-3 (GSK-3) is a ubiquitous serine-threonine protein kinase that participates in a multitude of cellular processes and has recently been implicated in the pathophysiology of a number of diseases. The aim of this study was to investigate the effects of GSK-3beta inhibition in a model of acute inflammation. Here, we have investigated the effects of TDZD-8, a potent and selective GSK-3beta inhibitor, in a mouse model of carrageenan-induced pleurisy. EXPERIMENTAL APPROACH: Injection of carrageenan into the pleural cavity of mice elicited an acute inflammatory response characterized by: accumulation of fluid containing a large number of neutrophils (PMNs) in the pleural cavity, infiltration of PMNs in lung tissues and subsequent lipid peroxidation, and increased production of nitrite/nitrate (NOx), prostaglandin E2 (PGE2), tumour necrosis factor-alpha, (TNF-alpha) and interleukin-1beta (IL-1beta). Furthermore, carrageenan induced an upregulation of the adhesion molecules ICAM-1 and P-selectin, iNOS, COX-2 as well as nitrotyrosine as determined by immunohistochemical analysis of lung tissues. KEY RESULTS: Administration of TDZD-8 (1, 3 or 10 mg kg(-1), i.p.), 30 min prior to injection of carrageenan, caused a dose-dependent reduction in all the parameters of inflammation measured. CONCLUSIONS AND IMPLICATIONS: Thus, based on these findings we propose that inhibitors of the activity of GSK-3beta, such as TDZD-8, may be useful in the treatment of various inflammatory diseases.


Subject(s)
Carrageenan/toxicity , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Lung/drug effects , Thiadiazoles/pharmacology , Animals , Apoptosis/drug effects , Cyclooxygenase 2/metabolism , Glycogen Synthase Kinase 3 beta , Immunohistochemistry , Lipid Peroxidation , Lung/cytology , Lung/enzymology , Male , Mice , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Peroxidase/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism
9.
Shock ; 26(2): 154-61, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16878023

ABSTRACT

We investigated whether (endogenous) hydrogen sulfide (H2S) protects the heart against myocardial ischemia and reperfusion injury. Furthermore, we investigated whether endogenous H2S is involved in the protection afforded by (1) ischemic preconditioning and (2) the second window of protection caused by endotoxin. The involvement of one of the potential (end) effectors of the cardioprotection afforded by H2S was investigated using the mitochondrial KATP channel blocker, 5-hydroxydecanoate (5-HD; 5 mg/kg). Animals were subjected to 25 min regional myocardial ischemia followed by reperfusion (2 h) and were pretreated with the H2S donor, sodium hydrosulfide (3 mg/kg i.v.). Animals were also subjected to shorter periods of myocardial ischemia (15 min) and reperfusion (2 h) and pretreated with an irreversible inhibitor of cystathionine-gamma-lyase, dl-propargylglycine (PAG; 50 mg/kg i.v.). Animals were also pretreated with PAG (50 mg/kg) and subjected to either (1) ischemic preconditioning or (2) endotoxin (1 mg/kg i.p.) 16 h before myocardial ischemia. Myocardial infarct size was determined by p-nitroblue tetrazolium staining. Administration of sodium hydrosulfide significantly reduced myocardial infarct size, and this effect was abolished by 5-HD. Administration of PAG (50 mg/kg) or 5-HD significantly increased infarct size caused by 15 min of myocardial ischemia. The delayed cardioprotection afforded by endotoxin was abolished by 5-HD or PAG. In contrast, PAG (50 mg/kg) did not affect the cardioprotective effects of ischemic preconditioning. These findings suggest that (1) endogenous H2S is produced by myocardial ischemia in sufficient amounts to limit myocardial injury and (2) the synthesis or formation of H2S by cystathionine-gamma-lyase may contribute to the second window of protection caused by endotoxin.


Subject(s)
Cardiotonic Agents/pharmacology , Hydrogen Sulfide/metabolism , Ischemic Preconditioning, Myocardial/methods , Myocardial Ischemia/prevention & control , Myocardial Reperfusion Injury/prevention & control , Alkynes/pharmacology , Animals , Cystathionine gamma-Lyase/antagonists & inhibitors , Decanoic Acids/pharmacology , Endotoxins/pharmacology , Enzyme Inhibitors/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , Hydroxy Acids/pharmacology , Male , Mitochondria/drug effects , Mitochondria/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Potassium Channel Blockers/pharmacology , Rats , Rats, Wistar , Sulfides/pharmacology
10.
Scand J Immunol ; 64(1): 61-8, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16784492

ABSTRACT

Previous studies have implicated a role of bacterial DNA, containing unmethylated cytosine-phosphate-guanosine (CpG) motifs, in the initiation of systemic inflammation. This is based on the ability of CpG-DNA to act in synergy with lipopolysaccharide (LPS) to trigger tumor necrosis factor alpha (TNFalpha) production in murine monocytes and to enhance LPS toxicity in rodents. In this study we investigated the capacity of CpG-DNA to trigger and modulate cytokine responses in human leukocytes. A human blood assay, as well as isolated cultures of monocytes and neutrophils, was exposed to the synthetic oligodeoxynucleotides (ODNs) CpG ODN (2006) and GpC ODN (2006-GC), alone or in combination with peptidoglycan or LPS. Plasma or supernatants were isolated and analyzed for TNFalpha, interleukin-1 beta (IL-1beta), IL-6 and IL-8 by ELISA. In the blood, 2006 (but not 2006-GC) induced the release of TNFalpha (P < 0.05) and possibly IL-1beta and IL-6. IL-8 was induced in a CpG-independent manner. When co-administered with peptidoglycan, both ODNs enhanced the release of cytokines, but not consistently CpG dependent. When co-administered with LPS, only IL-8 values were enhanced, whereas IL-6 was suppressed at early time points. In monocyte and neutrophil cultures, CpG dependent induction of cytokine release was not observed. However, both ODNs inhibited LPS-induced IL-6. In conclusion, the capacity of CpG DNA to trigger the release of TNFalpha and to enhance LPS-induced release of this cytokine is confirmed in human whole blood, but not in adherent human monocytes. Most effects of the ODNs on cytokine release in human leukocytes were CpG independent.


Subject(s)
CpG Islands/immunology , Cytokines/immunology , Leukocytes/immunology , Cytokines/blood , Cytokines/metabolism , Humans , Inflammation Mediators/immunology , Lipopolysaccharides/immunology , Monocytes/immunology , Neutrophils/immunology , Oligonucleotides/immunology , Peptidoglycan/immunology , Tumor Necrosis Factor-alpha/metabolism
11.
J Periodontal Res ; 41(3): 208-13, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16677290

ABSTRACT

BACKGROUND AND OBJECTIVE: Enamel matrix derivative (EMD), extracted from porcine tooth buds, has been shown to promote periodontal healing in patients with severe periodontitis. This involves modulation of the inflammatory response followed by the onset of periodontal regeneration. Based on these observations, we examined the ability of EMD to modulate the release of a pro-inflammatory cytokine [tumor necrosis factor (TNF)-alpha], an anti-inflammatory cytokine (interleukin-10) and a chemokine (interleukin- 8) in whole human blood challenged by bacterial cell wall components. MATERIAL AND METHODS: Whole blood from healthy donors was challenged by lipopolysaccharide or peptidoglycan and incubated with different concentrations of EMD or a cAMP analogue 8-(4-chlorophenyl)thio-cAMP (8-CPT-cAMP). TNF-alpha, interleukin-8 and interleukin-10 were analysed from plasma by enzyme-linked immunosorbent assay (ELISA) while cAMP levels of peripheral blood mononuclear cell lysates were analysed by enzyme immunoassay (EIA). RESULTS: We found that EMD attenuated the release of TNF-alpha and interleukin-8 in whole blood from healthy donors challenged by lipopolysaccharide or peptidoglycan, while the release of interleukin-10 was unchanged. Enamel matrix derivative also produced a four-fold increase in the cAMP levels of peripheral blood mononuclear cell lysates. Like EMD, 8-CPT-cAMP attenuated the formation of TNF-alpha, but not of interleukin-10, in blood challenged by lipopolysaccharide. CONCLUSION: Enamel matrix derivative limits the release of pro-inflammatory cytokines induced by lipopolysaccharide or peptidoglycan in human blood, suggesting that it has anti-inflammatory properties. We propose that this effect of EMD is, at least partly, secondary to an increase in the intracellular levels of cAMP in peripheral blood mononuclear cells.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dental Enamel Proteins/pharmacology , Animals , Cyclic AMP/analogs & derivatives , Cyclic AMP/blood , Cyclic AMP/pharmacology , Enzyme Inhibitors/pharmacology , Escherichia coli , Humans , Interleukin-10/blood , Interleukin-8/blood , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/enzymology , Lipopolysaccharides/pharmacology , Peptidoglycan/pharmacology , Staphylococcus aureus , Swine , Thionucleotides/pharmacology , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/drug effects
12.
Infect Immun ; 73(11): 7613-9, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16239565

ABSTRACT

Previous studies have indicated that peptidoglycan (PepG) from gram-positive bacteria can exert a priming effect on the innate immune response to lipopolysaccharide (LPS) from gram-negative bacteria. Here, we hypothesized that this priming effect may be preceded by enhanced expression of monocyte CD14, Toll-like receptor 2 (TLR2), and TLR4. In an ex vivo whole human blood model, we observed a substantial synergy between LPS and PepG in the release of tumor necrosis factor alpha and interleukin-1beta (IL-1beta) over the 24-h experimental period, whereas the effect on IL-8 and IL-10 release was more time dependent. The priming effect of PepG on cytokine release was preceded by a rapid upregulation of CD14, TLR2, and TLR4 expression on monocytes: at 3 hours there was a twofold increase in CD14 expression (P < 0.03), a fivefold increase in TLR2 expression (P < 0.03), and a twofold increase in TLR4 expression (P < 0.03). CD14 and TLR2 remained upregulated throughout the experimental period following exposure to PepG (P < 0.05). Only a transient upregulation of these monocyte receptors was observed following treatment with LPS or LPS plus PepG. In conclusion, the synergistic effect of LPS and PepG on cytokine release is preceded by a reciprocal upregulation of TLR2 and TLR4 by both bacterial cell wall components.


Subject(s)
Lipopolysaccharide Receptors/biosynthesis , Lipopolysaccharides/pharmacology , Monocytes/drug effects , Peptidoglycan/pharmacology , Signal Transduction/drug effects , Staphylococcus aureus/physiology , Toll-Like Receptor 2/biosynthesis , Toll-Like Receptor 4/biosynthesis , Cytokines/metabolism , Humans , Monocytes/metabolism , Up-Regulation/drug effects
13.
Eur Respir J ; 25(2): 225-34, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15684285

ABSTRACT

Thiazolidinedione rosiglitazone and 15-deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2), are two peroxisome proliferator-activated receptor (PPAR)-gamma ligands. The aim of this study was to investigate the effect of rosiglitazone and 15d-PGJ2 on the lung injury caused by bleomycin administration. Mice subjected to intratracheal administration of bleomycin developed significant lung injury. An increase in immunoreactivity to nitrotyrosine, poly(ADP ribose) polymerase (PARP) and inducible nitric oxide synthase as well as a significant loss of body weight and mortality was observed in the lung of bleomycin-treated mice. Administration of the two PPAR-gamma agonists rosiglitazone (10 mg x kg(-1) i.p.) and 15d-PGJ2 (30 microg x kg(-1) i.p.) significantly reduced the: 1) loss of body weight, 2) mortality rate, 3) infiltration of the lung with polymorphonuclear neutrophils (myeloperoxidase activity), 4) oedema formation, and 5) histological evidence of lung injury. Administration of rosiglitazone and 15d-PGJ2 also markedly reduced the nitrotyrosine, PARP and inducible nitric oxide synthase formation. In addition, treatment with the PPAR-gamma antagonist bisphenol A diglycidyl ether (1 mg x kg(-1) i.p. 30 min before the rosiglitazone or 15d-PGJ2) significantly antagonised the effect of the two PPAR-gamma agonists. These results demonstrate that the two peroxisome proliferator-activated receptor-gamma agonists, rosiglitazone and 15-deoxy-Delta12,14-prostaglandin J2, significantly reduce lung injury induced by bleomycin in mice.


Subject(s)
Prostaglandin D2/analogs & derivatives , Prostaglandin D2/pharmacology , Pulmonary Fibrosis/chemically induced , Thiazolidinediones/pharmacology , Tyrosine/analogs & derivatives , Analysis of Variance , Animals , Benzhydryl Compounds , Biopsy , Bleomycin , Epoxy Compounds/pharmacology , Immunoenzyme Techniques , Instillation, Drug , Male , Mice , Nitric Oxide Synthase/metabolism , Peroxidase/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Random Allocation , Rosiglitazone , Tyrosine/metabolism , Weight Loss/drug effects
14.
Transplant Proc ; 36(4): 849-53, 2004 May.
Article in English | MEDLINE | ID: mdl-15194291

ABSTRACT

Liver ischemia is of clinical interest because of its role in liver failure and also hepatic graft rejection. The generation of reactive oxygen species contributes to the injury that follows ischemia-reperfusion. One therapy utilizes the administration of antioxidants; however, only limited experience suggests a potential benefit of systemic administration of these compounds. To overcome the limitations of these compounds, small molecules with improved cell membrane permeability characteristics and higher potency, such as tempol, are being tested in vivo. Tempol, a membrane-permeable radical scavenger, interferes with the formation or the effects of many radicals, including superoxide anions, hydroxyl radicals, and peroxynitrite. The aim of this study was to investigate the effects of tempol in an in vivo rat model of liver ischemia-reperfusion injury. Male Wistar rats were pretreated with tempol (30 mg/kg, i.v.) 5 minutes prior to liver ischemia (for 30 minutes) and reperfusion (for 2 hours). The liver injury was assessed by measuring serum levels of transaminases, lactate dehydrogenase, and gamma-glutamyl transferase. Tempol significantly mitigated the increase in transaminases, lactate dehydrogenase, and gamma-glutamyl transferase following liver ischemia-reperfusion, suggesting an improvement in liver function and resistance to injury.


Subject(s)
Cyclic N-Oxides/pharmacology , Liver/blood supply , Reperfusion Injury/prevention & control , Animals , Disease Models, Animal , Free Radical Scavengers/pharmacology , Liver/drug effects , Liver/pathology , Liver Function Tests , Male , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Spin Labels
15.
Br J Pharmacol ; 141(1): 1-3, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14662733

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily of ligand-activated transcription factors that are related to retinoid, steroid and thyroid hormone receptors. The PPAR subfamily comprises of three members, PPAR-alpha, PPAR-beta and PPAR-gamma. There is good evidence that ligands of PPAR-gamma, including certain thiazolinediones, reduce myocardial tissue injury and infarct size. The use of PPAR-gamma agonists in the treatment of heart failure is, however, controversial.


Subject(s)
Heart Failure/drug therapy , PPAR gamma/therapeutic use , Animals , Disease Models, Animal , Humans , Ligands , Mice , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Pioglitazone , Rats , Thiazoles/pharmacology , Thiazoles/therapeutic use , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use
16.
Intensive Care Med ; 28(6): 775-88, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12107686

ABSTRACT

OBJECTIVE: To investigate the effects of tyrphostin AG 126, a tyrosine kinase inhibitor, on the multiple organ failure (MOF) caused by zymosan in the rat. DESIGN: Zymosan (500 mg/kg, suspended in saline solution, i.p.) causes an enhanced formation of reactive oxygen species, which contribute to the pathophysiology of MOF. After zymosan or saline administration, animals were monitored for 12 days. MEASUREMENTS AND RESULTS: Treatment of rats with tyrphostin AG 126 (10 mg/kg, 3 mg/kg or 1 mg/kg intraperitoneally, 1 h and 6 h after zymosan) attenuated the peritoneal exudation and the migration of polymorphonuclear cells caused by zymosan in a dose-dependent fashion. Tyrphostin AG 126 also attenuated the lung, liver, and intestinal injury (histology) as well as the increase in the levels of myeloperoxidase and malondialdehyde caused by zymosan in the lung, liver, and intestine. Immunohistochemical analysis for nitrotyrosine, poly (ADP-ribose) polymerase (PAR), iNOS, and COX-2 revealed a positive staining in lung, liver and intestine from zymosan-treated rats. The degree of staining for nitrotyrosine, PAR, iNOS, and COX-2 were markedly reduced in tissue sections obtained from zymosan-treated rats which had received tyrphostin AG 126. Furthermore, treatment of rats with tyrphostin AG 126 significantly reduced the production of peroxynitrite and of pro-inflammatory cytokines TNF-alpha and IL-1beta. CONCLUSIONS: This study provides the first evidence that the protein kinase inhibitor tyrphostin AG 126 attenuates the degree of MOF associated with zymosan-induced peritonitis in the rat.


Subject(s)
Enzyme Inhibitors/therapeutic use , Multiple Organ Failure/prevention & control , Protein-Tyrosine Kinases/antagonists & inhibitors , Tyrphostins/therapeutic use , Zymosan/toxicity , Animals , Male , Models, Animal , Multiple Organ Failure/chemically induced , Multiple Organ Failure/pathology , Nitric Oxide/blood , Peritonitis/chemically induced , Rats , Rats, Sprague-Dawley
17.
J Physiol Pharmacol ; 53(4 Pt 1): 555-69, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12512692

ABSTRACT

An enhanced formation of nitric oxide (NO) by the inducible NO synthase (iNOS) may contribute to the pathophysiology of hemorrhagic shock. This study investigates the effect of a novel, potent and selective inhibitor of iNOS activity (GW274150) on the circulatory failure and the organ injury and dysfunction associated with hemorrhagic shock in the anesthetised rat. Hemorrhage (sufficient to lower mean arterial blood pressure to 45 mmHg for 90 min) and subsequent resuscitation with shed blood resulted (within 4 h after resuscitation) in a delayed fall in blood pressure, renal and liver injury and dysfunction as well as the pancreatic injury. Pre-treatment of rats with GW274150 (5 mg/kg at 30 min prior to the onset of hemorrhage) attenuated the renal dysfunction as well as the liver and pancreatic injury caused by hemorrhage and resuscitation. Interestingly, GW274150 did not reduce the delayed fall in blood pressure associated with hemorrhagic shock. We propose that an enhanced formation of NO from iNOS contributes to the organ injury and dysfunction in hemorrhagic shock, and that highly selective inhibitors of iNOS activity, such as GW274150, may represent a novel therapeutic approach for the therapy of hemorrhagic shock.


Subject(s)
Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Shock, Hemorrhagic/pathology , Shock, Hemorrhagic/physiopathology , Sulfides/pharmacology , Tyrosine/analogs & derivatives , Animals , Blood Pressure/drug effects , Heart Rate/drug effects , Liver/drug effects , Liver/pathology , Liver/physiopathology , Lung/metabolism , Male , Multiple Organ Failure/pathology , Multiple Organ Failure/physiopathology , Neutrophils/pathology , Nitric Oxide Synthase Type II , Rats , Shock, Hemorrhagic/complications , Tyrosine/biosynthesis
18.
Eur J Pharmacol ; 430(1): 93-100, 2001 Oct 26.
Article in English | MEDLINE | ID: mdl-11698068

ABSTRACT

This study investigates the effects of a novel, water-soluble inhibitor of the activity of poly (adenosine 5'-diphosphate ribose) polymerase, 5-aminoisoquinolinone [5-aminoisoquinolin-1(2H)-one], on (i) poly (adenosine 5'-diphosphate ribose) polymerase activity in rat cardiac myoblasts and (ii) the infarct size caused by regional myocardial ischaemia and reperfusion in the rat. Exposure of H9c2 cells to hydrogen peroxide (H2O2, 1 mM) caused a significant increase in poly (adenosine 5'-diphosphate ribose) polymerase activity and an 80-90% reduction in mitochondrial respiration (cellular injury). Pretreatment of these cells with 5-aminoisoquinolinone (0.003-1 mM) caused a concentration-dependent inhibition of poly (adenosine 5'-diphosphate ribose) polymerase activity (IC50: approximately 4.5 microM, n=6-9) and cell injury (EC50: approximately 4.45 microM, n=9). In a rat model of myocardial infarction, left anterior descending coronary artery occlusion (25 min) and reperfusion (2 h) resulted in an infarct size of 50+/-3%. Administration (1 min before reperfusion) of 5-aminoisoquinolinone reduced myocardial infarct size in a dose-related fashion. Thus, 5-aminoisoquinolinone is a potent inhibitor of poly (adenosine 5'-diphosphate ribose) polymerase activity in cardiac myoblasts and reduces myocardial infarct size in vivo.


Subject(s)
Isoquinolines/pharmacology , Myocardial Ischemia/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Cells, Cultured , Enzyme Inhibitors/pharmacology , Heart/drug effects , Hydrogen Peroxide , Isoquinolines/chemical synthesis , Isoquinolines/therapeutic use , Myocardial Ischemia/pathology , Myocardial Reperfusion , Myocardium/metabolism , Myocardium/pathology , Nitroblue Tetrazolium , Rats , Staining and Labeling
19.
Shock ; 16(3): 178-82, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11531018

ABSTRACT

The pathophysiological mechanisms involved in mixed bacterial infections caused by gram-positive and gram-negative bacteria are largely unknown. The present study examines the potential interaction between lipopolysaccharide (LPS) and peptidoglycan (PepG) in the induction of the sepsis-associated cytokines tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), and IL-10 in whole human blood. Plasma values of these cytokines were measured by enzyme immunoassays and a TNF bioassay. Co-administration of PepG (10 microg/mL) or muramyl dipeptide (MDP, 1 microg/mL) with LPS (10 ng/mL) caused significantly elevated values of TNF-alpha and IL-6 in the blood that could not be obtained by the sum of the values obtained by each stimulant alone, or by 3-fold higher doses of either bacterial component alone. This phenomenon was observed 1 h after stimulation, throughout the experimental period (24 h), and with different doses of LPS and PepG. In contrast, the release of IL-10 was not influenced by the co-administration of PepG or MDP with LPS. The TNF-alpha release induced by co-administration of LPS and PepG was abrogated after pretreatment with a monoclonal antibody against CD14 (18D11). Addition of PepG or MDP to whole blood caused a 2-fold increase in the surface expression of CD14 on monocytes, as measured by flow cytometry. In contrast, LPS caused decreased expression of this receptor. Our data suggest that PepG and MDP primes human whole blood leukocytes for LPS-induced release of proinflammatory cytokines. We speculate that synergy between PepG and LPS may contribute to the pathogenesis in sepsis caused by mixed bacterial infections.


Subject(s)
Cytokines/blood , Lipopolysaccharides/pharmacology , Peptidoglycan/pharmacology , Sepsis/blood , Humans , Inflammation/blood , Interleukin-10/blood , Interleukin-6/blood , Lipopolysaccharide Receptors/blood , Lipopolysaccharide Receptors/drug effects , Lipopolysaccharides/administration & dosage , Peptidoglycan/administration & dosage , Tumor Necrosis Factor-alpha/metabolism
20.
FASEB J ; 15(11): 1941-52, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11532974

ABSTRACT

High density lipoproteins (HDLs) inhibit the cytokine-induced expression of endothelial cell adhesion molecules both in vitro and in vivo. We examined the ability of HDLs to mediate a functional anti-inflammatory effect by measuring their ability to prevent neutrophil adhesion and transmigration in vitro. Treatment of human endothelial cell cultures with physiologic concentrations of HDLs inhibited neutrophil binding by 68 +/- 5.9% (mean and se, n=6, P<0.05) and neutrophil transmigration by 48.7 +/- 6.7% (n=8, P<0.05). We then examined the effect of HDLs on inflammatory infiltration and subsequent multiple organ dysfunction syndrome (MODS), associated with trauma in a rat model of hemorrhagic shock. Rats given human HDLs (80 mg apo A-I/kg, i.v.) 90 min after hemorrhage (which reduced mean arterial pressure to 50 mmHg) and 1 min before resuscitation showed attenuation of the increases in the serum levels of markers of MODS normally observed in this model. Severe disruption of the architecture of tissues and the extensive cellular infiltration into those tissues were also largely inhibited in animals that received HDLs. Human HDLs attenuate the MODS associated with ischemia and reperfusion injury after hemorrhagic shock in rats.


Subject(s)
Lipoproteins, HDL/immunology , Multiple Organ Failure/immunology , Shock, Hemorrhagic/immunology , Adult , Animals , Biomarkers , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Chemokine CXCL2 , Chemokines/genetics , Disease Models, Animal , Endothelium, Vascular/cytology , Hemodynamics/drug effects , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Interleukin-8/metabolism , Kidney/physiopathology , Lipoproteins, HDL/pharmacology , Liver/injuries , Lung , Multiple Organ Failure/complications , Muscles/injuries , Neurons , Neutrophils/drug effects , Neutrophils/physiology , P-Selectin/genetics , P-Selectin/metabolism , Pancreas/injuries , RNA, Messenger/metabolism , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Shock, Hemorrhagic/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...