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 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
3.
J Exp Med ; 188(2): 305-15, 1998 Jul 20.
Article in English | MEDLINE | ID: mdl-9670043

ABSTRACT

The incidence of septic shock caused by gram-positive bacteria has risen markedly in the last few years. It is largely unclear how gram-positive bacteria (which do not contain endotoxin) cause shock and multiple organ failure. We have discovered recently that two cell wall fragments of the pathogenic gram-positive bacterium Staphylococcus aureus, lipoteichoic acid (LTA) and peptidoglycan (PepG), synergize to cause the induction of nitric oxide (NO) formation, shock, and organ injury in the rat. We report here that a specific fragment of PepG, N-acetylglucosamine-beta-[1--> 4]-N-acetylmuramyl-L-alanine-D-isoglutamine, is the moiety within the PepG polymer responsible for the synergism with LTA (or the cytokine interferon gamma) to induce NO formation in the murine macrophage cell line J774.2. However, this moiety is also present in the PepG of the nonpathogenic bacterium Bacillus subtilis. We have discovered subsequently that S. aureus LTA synergizes with PepG from either bacterium to cause enhanced NO formation, shock, and organ injury in the rat, whereas the LTA from B. subtilis does not synergize with PepG of either bacterium. Thus, we propose that the structure of LTA determines the ability of a particular bacterium to cause shock and multiple organ failure (pathogenicity), while PepG acts to amplify any response induced by LTA.


Subject(s)
Lipopolysaccharides/chemistry , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Macrophages/microbiology , Nitric Oxide Synthase/biosynthesis , Peptidoglycan/chemistry , Peptidoglycan/pharmacology , Staphylococcal Infections/metabolism , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Teichoic Acids/chemistry , Teichoic Acids/pharmacology , Animals , Bacillus subtilis/metabolism , Cell Line , Enzyme Induction/drug effects , Mice , Multiple Organ Failure/metabolism , Multiple Organ Failure/microbiology , Peptide Fragments/pharmacology , Rats , Shock, Septic/metabolism , Shock, Septic/microbiology , Staphylococcal Infections/microbiology
4.
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
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.
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
7.
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
8.
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.
Cancer Res ; 54(24): 6458-63, 1994 Dec 15.
Article in English | MEDLINE | ID: mdl-7987843

ABSTRACT

The nitric oxide synthase inhibitor NG-nitro-L-arginine (NOARG) was examined for its ability to alter energy metabolism in three murine tumors using 31P magnetic resonance spectroscopy. NOARG (10 mg/kg, i.v.) increased the inorganic phosphate:total phosphate ratio (Pi:total) 2-3-fold in the KHT, RIF-1, and SCCVII/Ha intradermal back tumors from 30 min to 6 h after injection, but the 31P magnetic resonance spectrum from normal tissue on the mouse back was unchanged after this treatment. NOARG (10 mg/kg, i.v.) injected 30 min before X-rays increased tumor cell survival 3-5-fold in SCCVII/Ha and 50-200-fold in RIF-1, measured using an in vivo/in vitro clonogenic assay. These effects were equivalent to those obtained from clamped tumors, indicating full radiobiological hypoxia. In KHT, only a 2-fold increase in radioresistance was observed after NOARG, which was less than the response of clamped tumors. In RIF-1 tumors, NOARG induced full radiobiological hypoxia when given from 30 min to 6 h prior to X-rays, consistent with the time course for the increase in Pi:total, measured by 31P magnetic resonance spectroscopy. Pi:total after NOARG doses of 0.1-10 mg/kg, i.v., increased in a dose-dependent manner in this tumor. Increased RIF-1 tumor radioresistance was similarly dependent on NOARG dose. The combination of the bioreductive agent RB6145 (300 mg/kg, i.p.) 15 min prior to NOARG (10 mg/kg, i.v.) produced greater than 5 decades of KHT tumor cell killing at 24 h after treatment. This combination also increased Pi:total 4.5-fold over the control value at 24 h in the KHT tumor. Histological examination of tumors at this time indicated extensive necrosis.


Subject(s)
Arginine/analogs & derivatives , Carcinoma/drug therapy , Energy Metabolism/drug effects , Sarcoma, Experimental/drug therapy , Animals , Arginine/pharmacology , Carcinoma/metabolism , Carcinoma/radiotherapy , Cell Hypoxia , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Female , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C3H , Nitroarginine , Nitroimidazoles/pharmacology , Phosphates/metabolism , Prodrugs/pharmacology , Sarcoma, Experimental/metabolism , Sarcoma, Experimental/radiotherapy
11.
Pharmacol Ther ; 87(1): 61-7, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10924742

ABSTRACT

Prostaglandins (PGs) and other eicosanoids are members of a large family of lipid mediators (autacoids). In 1978, Lefer and colleagues (Science 200, 52-55 [1978]) reported that prostacyclin reduces the myocardial tissue injury caused by coronary artery occlusion and reperfusion in the cat. Since this discovery, more than 50 papers have reported on the cardioprotective effects of vasodilator PGs, including prostacyclin. The cardioprotective effects of PGs are due in part to (1) a reduction in afterload, (2) an increase in coronary blood flow, (3) the inhibition of platelet function, and (4) the inhibition of the activation and extravasation of polymorphonuclear granulocytes. All of these effects are secondary to the activation of EP (E-type prostanoid)(2)-receptors, which activate G(s)-protein and, hence, adenylate cyclase. In addition, the protection of organs such as the heart by PGs has been attributed to a cytoprotective effect of these agents, the mechanism of which is largely unknown. We recently have discovered that certain E-type PGs, which do not activate EP(2)-receptors, also reduce myocardial infarct size, without causing a fall in blood pressure (EP(2)-receptor-mediated effects). Having provided a brief introduction into the role of eicosanoids in ischaemia-reperfusion injury of the heart, this review focuses on the recent discovery that selective agonists of EP(3)-receptors reduce myocardial infarct size, without causing haemodynamic side effects. The mechanisms of the cardioprotective effects of these agents are discussed, as are the therapeutic implications.


Subject(s)
Myocardial Infarction/prevention & control , Prostaglandins/therapeutic use , Receptors, Prostaglandin E/agonists , Animals , Humans , Receptors, Prostaglandin E, EP3 Subtype
12.
Pharmacol Ther ; 69(2): 143-51, 1996.
Article in English | MEDLINE | ID: mdl-8984508

ABSTRACT

Preconditioning of the heart with brief periods of ischaemia protects the myocardium for up to 90 min against a more sustained ischaemic injury. A "second window of protection" occurs 24 hr after preconditioning with ischaemia. The cardioprotective effects of ischaemic preconditioning involve the release of mediators (adenosine, bradykinin, catecholamines, prostaglandins, endothelin-1), which either alone or in concert activate protein kinase C, which translocates to the cell membrane. This manuscript reviews (i) the cardioprotective effects of ischaemic preconditioning, (ii) the underlying mechanisms, (iii) the effects of ischaemic preconditioning of other tissues (skeletal muscle, brain and kidney), and (iv) the clinical implications.


Subject(s)
Myocardial Ischemia/prevention & control , Protein Kinase C/pharmacology , Receptors, Adrenergic , Adaptation, Physiological , Organ Specificity
13.
Cardiovasc Res ; 31(6): 941-6, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8759250

ABSTRACT

OBJECTIVE: To investigate the effect of the ATP-sensitive potassium (KATP) channel inhibitor sodium 5-hydroxydecanoate (5-HD) on the reduction in myocardial infarct size afforded by ischaemic preconditioning in the sodium pentobarbitone rabbit. METHODS: New Zealand white rabbits were anaesthetised with sodium pentobarbitone and subjected to 60 min occlusion of the first antero-lateral branch of the left coronary artery (LAL) followed by 120 min reperfusion. Ischaemic preconditioning was achieved by a single episode of 5 min LAL occlusion followed by 15 min reperfusion prior to the 60 min occlusion. 5-HD (5 mg kg-1), an ischaemia selective KATP channel inhibitor, was administered into the left ventricle as a bolus injection 10 min prior to the onset of ischaemic preconditioning. Injection of Evans blue dye was used to determine the area of the left ventricle at risk and infarct size was determined by incubation of the area at risk with nitro-blue tetrazolium. RESULTS: There were no significant differences between groups in haemodynamics or area at risk. Ischaemic preconditioning resulted in a significant reduction in infarct size (27 +/- 8%) when compared to control animals (55 +/- 3%; P < 0.05). Pretreatment of animals with 5-HD completely abolished the cardioprotection seen with ischaemic preconditioning (50 +/- 6%). CONCLUSION: These results support the hypothesis that the cardioprotection afforded by ischaemic preconditioning in the pentabarbitone anaesthetised rabbit is dependent on the opening of KATP channels.


Subject(s)
Decanoic Acids/pharmacology , Hydroxy Acids/pharmacology , Myocardial Infarction/prevention & control , Myocardium/pathology , Potassium Channel Blockers , Animals , Myocardial Infarction/pathology , Myocardial Ischemia/pathology , Myocardial Reperfusion , Rabbits
14.
Cardiovasc Res ; 41(1): 126-34, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10325960

ABSTRACT

OBJECTIVE: Inhibitors of poly (ADP-ribose) synthetase (PARS) activity reduce the infarct size caused by regional myocardial ischaemia and reperfusion in the rabbit and rat in vivo. The mechanism of action of these inhibitors is unclear. Here we investigate the effects of the PARS inhibitor 3-aminobenzamide (3-AB) on infarct size caused by ischaemia and reperfusion of the isolated, perfused heart of the rat. We also investigate the role of PARS in the hydrogen peroxide-mediated cell injury/necrosis in rat cardiac myoblasts. METHODS: Rat isolated hearts perfused at constant pressure (80 mmHg) were subjected to 35 min of regional ischaemia and 2 h of reperfusion. Infarct size was determined at the end of the experiment using nitro-blue tetrazolium. 3-AB (300 microM) or 3-aminobenzoic acid (3-ABA, 300 microM) were infused during the reperfusion period. Rat cardiac myoblasts (H9c2 cells) were preincubated with the PARS inhibitors, 3-AB. nicotinamide (Nic) or 1,5-dihydroxyisoquinoline (ISO) or the inactive analogues 3-ABA or nicotinic acid (NicA) prior to exposure with hydrogen peroxide (1 mM). Cell injury was assessed by measuring mitochondrial respiration and cell necrosis by measuring the release of LDH. PARS activity was determined by measuring the incorporation of NAD into nuclear proteins. RESULTS: Regional ischaemia and reperfusion of the isolated rat heart resulted in an infarct size of 54% which was reduced by 3-AB, but not by 3-ABA. Exposure of rat cardiac myoblasts to hydrogen peroxide caused an increase in PARS activity and cell injury/necrosis which was attenuated by pretreatment with the PARS inhibitors. CONCLUSION: Inhibition of the activity of PARS attenuates the cell death associated with oxidant stress in rat cardiac myoblasts and heart.


Subject(s)
Benzamides/pharmacology , Enzyme Inhibitors/pharmacology , Heart/drug effects , Myocardial Reperfusion Injury/prevention & control , Poly(ADP-ribose) Polymerase Inhibitors , Aminobenzoates/pharmacology , Animals , Cell Death/drug effects , Cell Line , Hydrogen Peroxide , Isoquinolines/pharmacology , Male , Niacin/pharmacology , Niacinamide/pharmacology , Oxidative Stress , Perfusion , Poly(ADP-ribose) Polymerases/metabolism , Rats , Rats, Wistar , meta-Aminobenzoates
15.
Hypertension ; 31(6): 1240-7, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9622136

ABSTRACT

The peptide hormone relaxin (RLX) has been shown to elicit a powerful vasodilatory response in several target organs. This response is mediated by the stimulation of intrinsic nitric oxide (NO) generation. The present study was designed to clarify whether RLX directly promotes the relaxation of vascular smooth muscle cells through stimulation of NO generation. Vascular smooth muscle cells from bovine aortas were incubated with RLX at concentrations ranging from 1 nmol/L to 1 micromol/L. The expression and activity of NO synthase, production of NO, and the intracellular levels of cGMP and Ca2+ were determined. The cell morphology and signal transduction mechanisms of these bovine aortic smooth muscle cells in response to RLX were also studied. RLX stimulated the expression of immunoreactive inducible NO synthase and increased significantly and in a concentration-related fashion inducible NO synthase activity, NO generation, and intracellular cGMP levels. Concurrently, RLX significantly decreased cytosolic Ca2+ concentrations and caused changes in cell shape and the actin cytoskeleton that were consistent with cell relaxation. The signal transduction mechanisms leading to the enhanced expression of inducible NO synthase protein and activity caused by RLX involve the activation of tyrosine kinase, phosphatidylcholine-phospholipase C, and the transcription factor nuclear factor-kappaB, similar to bacterial endotoxins and proinflammatory cytokines. This study suggests that RLX is an endogenous agent capable of regulating vascular tone by activation of the L-arginine-NO pathway in vascular smooth muscle cells.


Subject(s)
Arginine/metabolism , Muscle, Smooth, Vascular/physiology , Nitric Oxide/metabolism , Relaxin/physiology , Analysis of Variance , Animals , Arginine/physiology , Calcium/metabolism , Cattle , Cells, Cultured , Enzyme Activation , Guanosine Monophosphate/metabolism , Immunohistochemistry , Microscopy, Electron , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide/physiology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Phosphatidylcholines/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Type C Phospholipases/metabolism
16.
Free Radic Biol Med ; 27(5-6): 493-503, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10490268

ABSTRACT

Reactive oxygen species (ROS) contribute to ischemia-reperfusion injury of the heart. This study investigates the effects of tempol, a membrane-permeable radical scavenger on (i) the infarct size caused by regional myocardial ischemia and reperfusion of the heart in vivo (rat, rabbit) and in vitro (rat), and (ii) the cell injury caused by hydrogen peroxide (H2O2) in rat cardiac myoblasts (H9c2 cells). In the anesthetized rat, tempol reduced the infarct size caused by regional myocardial ischemia (25 min) and reperfusion (2 h) from 60 +/- 3% (control, n = 8) to 24 +/- 5% (n = 6, p < .05). In the anesthetized rabbit, tempol also attenuated the infarct size caused by myocardial ischemia (45 min) and reperfusion (2 h) from 59 +/- 3% (control, n = 6) to 39 +/- 5% (n = 5, p < .05). Regional ischemia (35 min) and reperfusion (2 h) of the isolated, buffer-perfused heart of the rat resulted in an infarct size of 54 +/- 4% (control n = 7). Reperfusion of hearts with buffer containing tempol (n = 6) caused a 37% reduction in infarct size (n = 6, p < .05). Pretreatment of rat cardiac myoblasts with tempol attenuated the impairment in mitochondrial respiration caused by H2O2 (1 mM for 4 h). Thus, the membrane-permeable radical scavenger tempol reduces myocardial infarct size in rodents.


Subject(s)
Cyclic N-Oxides/pharmacology , Free Radical Scavengers/pharmacology , Myocardial Infarction/drug therapy , Myocardial Ischemia/drug therapy , Animals , Blood Flow Velocity/drug effects , Blood Pressure/drug effects , Cell Line , Disease Models, Animal , Heart Rate/drug effects , Hydrogen Peroxide/pharmacology , Malondialdehyde/metabolism , Mitochondria, Heart/drug effects , Myocardial Infarction/pathology , Myocardial Reperfusion , Oxidative Stress/drug effects , Rabbits , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Spin Labels
17.
Int J Radiat Oncol Biol Phys ; 29(3): 443-7, 1994 Jun 15.
Article in English | MEDLINE | ID: mdl-7516319

ABSTRACT

PURPOSE: To determine the effects of the nitric oxide synthase inhibitor, nitro-L-arginine on energy metabolism in transplantable and spontaneous murine tumors. METHODS AND MATERIALS: The responses of the transplantable murine tumor SCCVII/Ha and a range of spontaneously arising murine mammary adenocarcinomas to 10 mg/kg IV nitro-L-arginine were examined using in vivo 31P magnetic resonance spectroscopy (MRS). The influence of Hypnorm/Hypnovel anesthesia on the response to nitro-L-arginine was also determined in the SCCVII/Ha tumors. Data were expressed as changes in the inorganic phosphate peak area relative to the sum of all peak areas from the 31P MR spectrum, or Pi/total. RESULTS: Nitro-L-arginine at 10 mg/kg IV increased Pi/total 2-3-fold in the SCCVII/Ha tumors for at least 2 h after administration, in both anesthetized and nonanesthetized mice, consistent with increased tumor hypoxia. Similar increases in Pi/total were observed after 10 mg/kg IV nitro-L-arginine in 13 spontaneous murine tumors from three different mouse strains, where anesthetic was used. CONCLUSION: The results indicate that tumor metabolism may be modified by an inhibitor of nitric oxide synthesis, that this modification occurs in both transplantable and spontaneous murine tumors and is not affected by anesthetic.


Subject(s)
Amino Acid Oxidoreductases/antagonists & inhibitors , Arginine/analogs & derivatives , Carcinoma, Squamous Cell/metabolism , Mammary Neoplasms, Animal/metabolism , Animals , Arginine/pharmacology , Mice , Mice, Inbred C3H , Neoplasm Transplantation , Nitric Oxide Synthase , Nitroarginine
18.
Proc Biol Sci ; 241(1302): 195-200, 1990 Sep 22.
Article in English | MEDLINE | ID: mdl-1979444

ABSTRACT

The stimulation of gastric-acid secretion by pentagastrin, a synthetic analogue of the endogenous peptide gastrin, is associated with an increased blood flow to the stomach mucosa, commonly referred to as functional hyperaemia. There are at least two potent vasodilator substances, the local release of which from endothelial cells could contribute to this hyperaemia, endothelium-derived relaxing factor (EDRF) and prostacyclin. EDRF has been identified as nitric oxide, released enzymatically from the guanidino group of L-arginine. In the present studies, the involvement of prostacyclin in the pentagastrin-induced increase in stomach blood flow was eliminated by using the cyclo-oxygenase inhibitor indomethacin. Thus this work was designed to elucidate the participation of EDRF/NO in the pentagastrin-induced hyperaemia and not its relative importance to prostacyclin. The increase in blood flow to the gastric mucosa in response to pentagastrin was measured by laser Doppler flowmetry in situ. Inhibition of EDRF/NO biosynthesis with the L-arginine analogues NG-monomethyl-L-arginine (MeArg) or N omega-nitro-L-arginine (NO2Arg) significantly attenuated (by more than 80%) the increase in mucosal blood flow in response to pentagastrin. However, infusions of the natural substrate L-arginine reversed the inhibitor effect of MeArg on pentagastrin-induced increase in mucosal blood flow. Local intra-arterial injections of the endothelium-independent vasodilator glyceryl trinitrate produced a dose-related increase in blood flow to the rat stomach mucosa that was unaffected by infusion of MeArg. Thus, in the absence of prostacyclin, EDRF/NO participates in the pentagastrin-induced increase in blood flow to the rat stomach mucosa.


Subject(s)
Blood Pressure/physiology , Intestinal Mucosa/blood supply , Nitric Oxide/physiology , Pentagastrin/pharmacology , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Indomethacin/pharmacology , Male , Rats , Rats, Inbred Strains , Reference Values , Regional Blood Flow/drug effects , omega-N-Methylarginine
19.
Proc Biol Sci ; 253(1338): 233-8, 1993 Sep 22.
Article in English | MEDLINE | ID: mdl-7694300

ABSTRACT

Bacterial lipopolysaccharide (LPS) is the toxic moiety of the gram-negative bacterial outer membrane which is responsible for many of the pathophysiological events that occur during endotoxic shock. Here we investigate the hypothesis that endogenous glucocorticoids modulate the formation of nitric oxide (NO) and of vasodilator cyclooxygenase metabolites in response to LPS. Intravenous administration of a small dose of Escherichia coli LPS (0.1 mg kg-1) to normal Wistar rats caused a moderate fall in blood pressure, and 120 min of endotoxaemia was not associated with an attenuation of the rise in blood pressure elicited by intravenous injection of noradrenaline (NA; vascular hyporeactivity). When adrenalectomized (ADX) rats, which lack endogenous glucocorticoids, were subjected to the same dose of LPS, they developed a much more severe form of circulatory shock, which was characterized by a profound fall in blood pressure and a vascular hyporeactivity to NA. Both hypotension and vascular hyporeactivity were prevented by pre-treatment with dexamethasone. Inhibition of NO biosynthesis with NG-methyl-L-arginine significantly attenuated the hypotension and the vascular hyporeactivity to NA caused by LPS in ADX rats. Similarly, the cyclooxygenase inhibitor indomethacin significantly attenuated the circulatory failure elicited by LPS in the ADX rats. Interestingly, 120 min of endotoxaemia resulted in a de novo biosynthesis of an induced isoform of NO synthase in the lungs of ADX, but not normal Wistar, rats. This induction of NO synthase was prevented by dexamethasone pre-treatment.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Amino Acid Oxidoreductases/metabolism , Arginine/analogs & derivatives , Blood Pressure/drug effects , Dexamethasone/pharmacology , Glucocorticoids/physiology , Indomethacin/pharmacology , Lipopolysaccharides/toxicity , Nitric Oxide/biosynthesis , Prostaglandin-Endoperoxide Synthases/metabolism , Adrenalectomy , Animals , Arginine/pharmacology , Calcium/pharmacology , Escherichia coli , Male , Nitric Oxide Synthase , Norepinephrine/pharmacology , Rats , Rats, Wistar , Shock, Septic/physiopathology , Time Factors , omega-N-Methylarginine
20.
Am J Cardiol ; 56(15): 978-82, 1985 Dec 01.
Article in English | MEDLINE | ID: mdl-3907318

ABSTRACT

Defibrotide, a partially depolymerized polydeoxyribonucleotide obtained from mammalian lungs, was found to stimulate prostacyclin (PGI2) production and to possess significant antithrombotic and fibrinolytic activities. The present study was designed to evaluate the actions of defibrotide on feline myocardial ischemia, produced by 3 hours of occlusion of the left anterior descending coronary artery (LAD) and followed by 2 hours of reperfusion. Intravenous administration of defibrotide (32 mg/kg/hour subsequent to a 32 mg/kg bolus injection), beginning 30 minutes after LAD occlusion, resulted in a 60% reduction in loss of CK specific activity from ischemic myocardium at 5 hours, while the nonischemic myocardium remained unaffected. Defibrotide largely antagonized the increase in ST segment during LAD occlusion and prevented the appearance of a Q wave during early reperfusion, which was found in all vehicle-treated cats. Although 2 of 8 vehicle-treated cats died from ventricular fibrillation and another had severe ventricular tachyarrhythmia, none of the defibrotide-treated cats had similar severe changes in the electrocardiogram and all 7 cats survived the 5-hour observation period. Defibrotide had no direct action on general hemodynamic functions. In a separate set of experiments, defibrotide (0.1 mg/ml) produced an 8- to 10-fold stimulation of PGI2 release. The data suggest a remarkable protective potential of defibrotide on reperfusion damage of the ischemic myocardium, which may be associated with a PGI2-related mechanism.


Subject(s)
Coronary Disease/physiopathology , Fibrinolytic Agents/pharmacology , Heart/drug effects , Polydeoxyribonucleotides/pharmacology , Adenosine Triphosphate/metabolism , Animals , Blood Platelets/metabolism , Cats , Creatine Kinase/analysis , Electrocardiography , Epoprostenol/biosynthesis , Female , Hemodynamics/drug effects , Male , Myocardium/metabolism , Platelet Aggregation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL