Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
Add more filters










Publication year range
1.
Leukemia ; 27(10): 2040-2046, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23538752

ABSTRACT

Hematopoietic stem/progenitor cells (HSPCs) function to give rise to mature blood cells. Effective DNA damage response (DDR) and maintenance of genomic stability are crucial for normal functioning of HSPCs. Mammalian target of rapamycin (mTOR) integrates signals from nutrients and growth factors to control protein synthesis, cell growth, survival and metabolism, and has been shown to regulate DDR in yeast and human cancer cells through the p53/p21 signaling cascade. Here, we show that gene targeting of mTOR in HSPCs causes a defective DDR due to a variety of DNA damage agents, mimicking that caused by deficient FANCD2, a key component of the Fanconi anemia (FA) DDR machinery. Mechanistically, mTOR(-/-) HSPCs express drastically reduced FANCD2. Consistent with these genetic findings, inactivation of mTOR in human lymphoblast cells by pp242 or Torin 1, mTOR kinase inhibitors, suppresses FANCD2 expression and causes a defective DDR that can be rescued by reconstitution of exogenous FANCD2. Further mechanistic studies show that mTOR deficiency or inactivation increases phosphorylation and nuclear translocation of nuclear factor (NF)-κB, which results in an enhanced NF-κB binding to FANCD2 promoter to suppress FANCD2 expression. Thus, mTOR regulates DDR and genomic stability in hematopoietic cells through a noncanonical pathway involving NF-κB-mediated FANCD2 expression.


Subject(s)
DNA Damage , Fanconi Anemia Complementation Group D2 Protein/physiology , Hematopoietic Stem Cells/pathology , Lymphocytes/pathology , NF-kappa B/metabolism , TOR Serine-Threonine Kinases/physiology , Animals , Apoptosis , Blotting, Western , Cells, Cultured , Chromosome Breakage , Comet Assay , Electrophoretic Mobility Shift Assay , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fluorescent Antibody Technique , Genomic Instability , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Lymphocytes/drug effects , Lymphocytes/metabolism , Mice , Mice, Knockout , NF-kappa B/genetics , Naphthyridines/pharmacology , Peptide Fragments/pharmacology , Phosphorylation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
2.
J Endotoxin Res ; 8(2): 99-107, 2002.
Article in English | MEDLINE | ID: mdl-12028749

ABSTRACT

Recent in vitro studies have shown that isohelenin, a sesquiterpene lactone, inhibits the NF-kappaB pathway. This study examines the effect of isohelenin in endotoxic shock induced by administration of Escherichia coli endotoxin in male Wistar rats. A group of rats received isohelenin (2 mg/kg intraperitoneally) 15 min before endotoxin. In vehicle-treated rats, administration of endotoxin caused severe hypotension, which was associated with a marked hyporeactivity to norepinephrine and acetylcholine in ex vivo aortas. Elevated levels of plasma nitrate/nitrite, metabolites of nitric oxide (NO), were also found. These inflammatory events were preceded by cytosolic degradation of inhibitor-kappaBalpha (IkappaBalpha) and activation of nuclear factor-kappaB (NF-kappaB) in the lung within 15 min of endotoxin administration. Treatment with isohelenin resulted in hemodynamic improvement and reduced plasma levels of NO metabolites. Nuclear translocation of NF-kappaB was inhibited by isohelenin treatment in the lung, whereas degradation of IkappaBalpha was unchanged. In a separate set of experiments, treatment with isohelenin significantly improved survival in mice challenged with endotoxin. We conclude that isohelenin exerts beneficial therapeutic effects during endotoxic shock through inhibition of NF-kappaB.


Subject(s)
Endotoxins/toxicity , Lipopolysaccharides/toxicity , NF-kappa B/antagonists & inhibitors , Sesquiterpenes/therapeutic use , Shock, Septic/prevention & control , Acetylcholine/metabolism , Animals , Blotting, Western , Electrophoresis , Hemodynamics/drug effects , Lung/metabolism , Mice , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Nitrates/metabolism , Nitrites/metabolism , Norepinephrine/metabolism , Proteins/metabolism , Rats , Shock, Septic/physiopathology
3.
Gut ; 48(5): 610-22, 2001 May.
Article in English | MEDLINE | ID: mdl-11302957

ABSTRACT

BACKGROUND: Interleukin 10 (IL-10) exerts a wide spectrum of regulatory activities in immune and inflammatory responses. AIMS: The aim of this study was to investigate the role of endogenous IL-10 on modulation of the early inflammatory response after splanchnic ischaemia and reperfusion. METHODS: Intestinal damage was induced by clamping the superior mesenteric artery and the coeliac trunk for 45 minutes followed by reperfusion in IL-10 deficient mice (IL-10(-/-)) and wild-type controls. RESULTS: IL-10(-/-) mice experienced a higher rate of mortality and more severe tissue injury compared with wild-type mice subjected to ischaemia and reperfusion. Splanchnic injury was characterised by massive epithelial haemorrhagic necrosis, upregulation of P-selectin and intercellular adhesion molecule 1, and neutrophil infiltration. The degree of oxidative and nitrosative damage was significantly higher in IL-10(-/-) mice than in wild-type littermates, as indicated by elevated malondialdehyde levels and formation of nitrotyrosine. Plasma levels of the proinflammatory cytokines tumour necrosis factor alpha and interleukin 6 were also greatly enhanced in comparison with wild-type mice. These events were preceded by increased immunostaining and activity of the stress regulated c-Jun NH(2) terminal kinase and activation of the transcription factor activator protein 1 in the cellular nuclei of damaged tissue. CONCLUSIONS: These data demonstrate that endogenous IL-10 exerts an anti-inflammatory role during reperfusion injury, possibly by regulating early stress related genetic response, adhesion molecule expression, neutrophil recruitment, and subsequent cytokine and oxidant generation.


Subject(s)
Acute-Phase Reaction/physiopathology , Interleukin-10/deficiency , Mesentery/blood supply , Reperfusion Injury/physiopathology , Splanchnic Circulation/physiology , Acute-Phase Reaction/etiology , Animals , Intercellular Adhesion Molecule-1/physiology , Interleukin-6/physiology , JNK Mitogen-Activated Protein Kinases , Malondialdehyde/pharmacology , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/physiology , Neutrophil Infiltration/physiology , P-Selectin/physiology , Reperfusion Injury/complications , Severity of Illness Index , Transcription Factor AP-1/physiology , Tumor Necrosis Factor-alpha/physiology , Up-Regulation
4.
Brain Res ; 881(1): 88-97, 2000 Oct 20.
Article in English | MEDLINE | ID: mdl-11033098

ABSTRACT

Brain injury due to bacterial meningitis results in a high mortality rate and significant neurologic sequelae in survivors. The objective of this study was to determine if the application of moderate hypothermia shortly after the administration of antibiotics would attenuate the inflammatory response and increase in intracranial pressure that occurs in meningitis. For this study we used a rabbit model of severe Group B streptococcal meningitis. The first component of this study evaluated the effects of hypothermia on blood-brain barrier function and markers of inflammation in meningitic animals. The second part of the study evaluated the effects of hypothermia on intracranial pressure, cerebral perfusion pressure and brain edema. This study demonstrates that the use of hypothermia preserves CSF/serum glucose ratio, decreases CSF protein and nitric oxide and attenuates myeloperoxidase activity in brain tissue. In the second part of this study we show a decrease in intracranial pressure, an improvement in cerebral perfusion pressure and a decrease in cerebral edema in hypothermic meningitic animals. We conclude that in the treatment of severe bacterial meningitis, the application of moderate hypothermia initiated shortly after antibiotic therapy improves short-term physiologic measures associated with brain injury.


Subject(s)
Blood-Brain Barrier/physiology , Hypothermia, Induced , Meningitis, Bacterial/therapy , Streptococcal Infections/therapy , Streptococcus agalactiae , Animals , Blood Pressure/physiology , Body Temperature/physiology , Hypothermia, Induced/methods , Intracranial Pressure/physiology , Male , Meningitis, Bacterial/cerebrospinal fluid , Meningitis, Bacterial/physiopathology , Nitric Oxide/cerebrospinal fluid , Peroxidase/metabolism , Rabbits , Subarachnoid Space/pathology
5.
Life Sci ; 67(4): 365-72, 2000 Jun 16.
Article in English | MEDLINE | ID: mdl-11003046

ABSTRACT

Reactive oxygen and nitrogen species participate in the inflammatory process during meningitis. Among them, superoxide, nitric oxide (NO), and their reaction product peroxynitrite exert cytotoxic effects. Mercaptoethylguanidine (MEG) exerts beneficial effects in in vivo inflammatory conditions by scavenging peroxynitrite and inhibiting the inducible NO synthase. This study was designed to investigate whether MEG may attenuate inflammation and brain injury in experimental meningitis. Meningitis increased nitrite/nitrate, and protein content in the cerebrospinal fluid (CSF). In the brain tissue high levels of malondialdehyde and formation of nitrotyrosine indicated lipid peroxidation and nitrosative stress, respectively. Myeloperoxidase activity was increased indicating accumulation of neutrophils into the brain parenchyma. Treatment with MEG decreased nitrite/nitrate levels whereas it did not affect the bacterial clearance from the CSF. Furthermore, treatment with MEG markedly reduced brain tissue levels of myeloperoxidase and malondialdehyde. These data demonstrate that MEG could have a therapeutic role in meningitis.


Subject(s)
Enzyme Inhibitors/therapeutic use , Guanidines/therapeutic use , Meningitis, Bacterial/drug therapy , Nitric Oxide Synthase/antagonists & inhibitors , Streptococcal Infections/drug therapy , Tyrosine/analogs & derivatives , Animals , Brain/drug effects , Brain/metabolism , Immunoenzyme Techniques , Male , Malondialdehyde/metabolism , Meningitis, Bacterial/cerebrospinal fluid , Meningitis, Bacterial/microbiology , Meningitis, Meningococcal , Nitrates/cerebrospinal fluid , Nitric Oxide/metabolism , Nitrites/cerebrospinal fluid , Peroxidase/metabolism , Proteins/analysis , Rabbits , Streptococcal Infections/cerebrospinal fluid , Streptococcal Infections/microbiology , Streptococcus agalactiae , Tyrosine/metabolism
6.
Shock ; 14(2): 134-41, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10947156

ABSTRACT

Peroxynitrite-mediated DNA strand breaks trigger poly (ADP-ribose) synthetase (PARS) activation, resulting in intracellular energetic failure and organ dysfunction. We investigated the role of PARS activation on the inflammatory and functional response of the intestine to mesenteric ischemia-reperfusion injury. Anesthetized rats exposed to 15 min occlusion of the superior mesenteric artery showed an increased mucosal PARS activity (ex vivo incorporation of radiolabelled NAD+ in gut mucosal scrapings) as soon as 10 min after reperfusion. During the first 30 min of reperfusion, significant mucosal damage developed, as well as mucosal hyperpermeability to a 4000 MW fluorescent dextran (FD4). These alterations were significantly reduced by treatment with the NO synthase inhibitor L-NMA, which blocks the production of peroxynitrite, as well as with the PARS inhibitors 3-aminobenzamide and nicotinamide, whereas they were markedly enhanced by the glutathione depletor L-buthionine-(S,R)-sulfoximine. Also, PARS inhibition significantly reduced ileal neutrophil infiltration (myeloperoxidase activity) at 3 h reperfusion. In a second set of experiments, the effects of 15 or 30 min ischemia followed by 3 h reperfusion were evaluated in PARS knockout and wild-type mice. Significant protection against histological damage, neutrophil infiltration, and mucosal barrier failure (evaluated by the mucosal-to-serosal FD4 clearance of everted ileal sacs incubated ex vivo) was noted in PARS knockout mice, who also showed reduced alterations in remote organs, as shown by lesser lipid peroxidation (malondialdehyde formation) and neutrophil infiltration in the lung and liver. In conclusion, PARS plays a crucial role in mediating intestinal injury and dysfunction in the early and late phases of mesenteric reperfusion. Pharmacological inhibition of PARS may be a novel approach to protect tissues from reperfusion-related damage.


Subject(s)
Intestinal Mucosa/enzymology , Ischemia/enzymology , Mesentery/blood supply , Nitrates/metabolism , Poly(ADP-ribose) Polymerases/physiology , Animals , Arterial Occlusive Diseases/complications , Arterial Occlusive Diseases/physiopathology , Chemotaxis, Leukocyte , DNA Damage , Energy Metabolism , Enzyme Induction , Enzyme Inhibitors/pharmacology , Ileum/metabolism , Intestinal Mucosa/physiopathology , Ischemia/physiopathology , Lipid Peroxidation/drug effects , Liver/pathology , Lung/pathology , Male , Mesenteric Artery, Superior , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Neutrophils/immunology , Nitric Oxide Synthase/antagonists & inhibitors , Oxidative Stress , Peroxidase/metabolism , Poly(ADP-ribose) Polymerases/deficiency , Poly(ADP-ribose) Polymerases/genetics , Rats , Rats, Wistar , Reperfusion Injury/enzymology , omega-N-Methylarginine/pharmacology
7.
Nitric Oxide ; 4(4): 412-22, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10944426

ABSTRACT

Acute lung injury (ALI) is associated with diminished surfactant activity and pulmonary hypertension. NONOates are soluble NO donors which release NO in solution. Intratracheal NONOates reduce pulmonary hypertension and improve oxygenation in ALI. We hypothesized that the pharmacologic properties of NO donors would be unaltered after surfactant admixture in vitro and that aerosolized NONOate activity would be enhanced by surfactant pretreatment in vivo. NO donors were added to saline or surfactant and analyzed for nitrite/nitrate production and aortic ring vasodilation. Surfactant did not alter nitrate/nitrite production or aortic ring vasodilation. A porcine model of ALI with pulmonary hypertension was produced using intravenous oleic acid. Animals were assigned to Surfactant-Saline, Surfactant-NONOate, Saline-Saline, or Saline-NONOate groups. Saline or surfactant was instilled into the trachea, followed by gas exchange, pulmonary function, and hemodynamic measurements. NONOate or saline was then aerosolized, and additional data were collected. Oxygenation was improved in the Surfactant-NONOate group, while pulmonary hypertension was selectively reduced in both NONOate groups. Aerosolized NONOate following surfactant pretreatment improves oxygenation and reduces pulmonary hypertension in ALI.


Subject(s)
Hypertension, Pulmonary/drug therapy , Lung/physiopathology , Nitric Oxide Donors/pharmacology , Penicillamine/analogs & derivatives , Pulmonary Gas Exchange/drug effects , Pulmonary Surfactants/pharmacology , Respiratory Distress Syndrome/drug therapy , Amino Acids, Diamino/pharmacology , Animals , Aorta/drug effects , Aorta/physiology , Disease Models, Animal , Drug Synergism , Hemodynamics/drug effects , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , In Vitro Techniques , Lung/drug effects , Lung/pathology , Male , Methemoglobin/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Nitric Oxide/pharmacology , Oleic Acid , Penicillamine/pharmacology , Random Allocation , Rats , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/physiopathology , S-Nitroso-N-Acetylpenicillamine , Swine
8.
Circulation ; 101(9): 1019-26, 2000 Mar 07.
Article in English | MEDLINE | ID: mdl-10704170

ABSTRACT

BACKGROUND: The anti-inflammatory cytokine interleukin-10 (IL-10) has been detected in the plasma of patients with myocardial ischemia/reperfusion. The aim of our study was to investigate the role of endogenously produced IL-10 in myocardial ischemia/reperfusion. METHODS AND RESULTS: In the present study, we used wild-type and IL-10-deficient mice subjected to myocardial ischemia/reperfusion. Significant levels of IL-10 were produced in wild-type mice at 2 to 6 hours after myocardial reperfusion. The genetic deletion of IL-10 enhanced neutrophil infiltration into the reperfused tissues at 6 hours after reperfusion and increased infarct size and myocardial necrosis. Furthermore, in the absence of IL-10, an enhancement of the inflammatory response was seen, as demonstrated by increased plasma levels of tumor necrosis factor-alpha, nitrite/nitrate (breakdown products of NO), and increased tissue expression of intercellular adhesion molecule-1. Reperfusion for 24 hours was associated with a 75% mortality rate in IL-10-deficient mice, whereas no deaths occurred in the wild-type animals. CONCLUSIONS: The present findings provide the first direct evidence that endogenous IL-10 inhibits the production of tumor necrosis factor-alpha and NO and serves to protect the ischemic and reperfused myocardium through the suppression of neutrophil recruitment.


Subject(s)
Coronary Disease/physiopathology , Interleukin-10/biosynthesis , Myocardial Infarction/pathology , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Neutrophils/physiology , Animals , Cell Movement/physiology , Hemodynamics , Intercellular Adhesion Molecule-1/metabolism , Interleukin-10/physiology , Male , Mice , Mice, Inbred C57BL , Myocardial Ischemia/mortality , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/mortality , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Nitric Oxide/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis
9.
Shock ; 13(2): 126-34, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10670842

ABSTRACT

Expression of the inducible isoform of nitric oxide (NO) synthase, and the formation of peroxynitrite from NO and superoxide are responsible for some of the pathophysiological alterations seen during reperfusion injury and in various inflammatory conditions. Some of the effects of peroxynitrite are related to DNA single-strand breakage, and activation of poly (ADP-ribose) synthetase. Here we investigated the effect of nicaraven (2(R,S)-1,2-bis(nicotinamido)propane), a known hydroxyl radical scavenger compound and neuroprotective agent, on several NO- and peroxynitrite related pathways in vitro, and in shock and inflammation in vivo. Nicaraven, at 10 microM-10 mM, failed to inhibit the peroxynitrite-induced oxidation of dihydrorhodamine 123, indicating that the agent does not act as a scavenger of peroxynitrite. In RAW murine macrophages stimulated with peroxynitrite, nicaraven caused a dose-dependent, slight inhibition of poly (ADP-ribose) synthetase activation, possibly due to a direct inhibitory effect on the catalytic activity of poly (ADP-ribose) synthetase. Nicaraven partially protected against the peroxynitrite-induced suppression of mitochondrial respiration in RAW macrophages and caused a slight, dose-dependent inhibition of nitrite production in RAW macrophages stimulated with bacterial lipopolysaccharide. We next investigated the effect of nicaraven treatment in a variety of models of inflammation and reperfusion injury. Nicaraven (at 10-100 microg/paw) exerted significant protective effects in the carrageenan-induced paw edema model and (at 100 mg/kg i.v.) reduced neutrophil infiltration and histological damage in splanchnic artery occlusion-reperfusion injury. However, nicaraven failed to alter the course of hemorrhagic and endotoxic shock and arthritis in rodent models. The current data indicate the limited role of hydroxyl radicals in the pathogenesis of the inflammatory conditions tested.


Subject(s)
Free Radical Scavengers/pharmacology , Inflammation/metabolism , Niacinamide/analogs & derivatives , Nitric Oxide/metabolism , Shock, Hemorrhagic/metabolism , Shock, Septic/metabolism , Animals , Antioxidants/pharmacology , Arthritis, Experimental/chemically induced , Arthritis, Experimental/drug therapy , Carrageenan , Dose-Response Relationship, Drug , Edema/chemically induced , Edema/drug therapy , Enzyme Activation/drug effects , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Male , Mice , Mice, Inbred DBA , Niacinamide/pharmacology , Nitrates/pharmacology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Oxidation-Reduction/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Rats , Rats, Wistar , Reperfusion Injury/metabolism , Splanchnic Circulation/drug effects
10.
Shock ; 13(1): 60-6, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10638671

ABSTRACT

The nuclear enzyme poly (ADP ribose) synthetase (PARS) has been shown to play an important role in the pathogenesis of various forms of ischemia or reperfusion injury and circulatory shock. Recent studies demonstrated that inhibition or genetic inactivation of PARS is beneficial in the early phase of myocardial reperfusion injury. The aim of the present study was to investigate whether inactivation of PARS influences the delayed myocardial necrosis and the production of the proinflammatory cytokine tumor necrosis factor alpha (TNFalpha), the anti-inflammatory cytokine interleukin-10 (IL-10), and the free radical nitric oxide in the late stage of myocardial reperfusion injury. The results demonstrate that genetic disruption of PARS provides marked protection against the delayed myocardial ischemia and reperfusion injury. In addition, in the absence of functional PARS, a suppression of TNFalpha, IL-10, and nitric oxide production was found. These findings provide direct evidence that PARS activation participates in the development of delayed cell injury and delayed mediator production in myocardial reperfusion injury.


Subject(s)
Hemodynamics/physiology , Interleukin-10/biosynthesis , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/pathology , Poly(ADP-ribose) Polymerases/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Blood Pressure , Heart Rate , Mice , Mice, Knockout , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Necrosis , Peroxidase/metabolism , Phosphocreatine/blood , Poly(ADP-ribose) Polymerases/deficiency , Poly(ADP-ribose) Polymerases/genetics
11.
Am J Respir Crit Care Med ; 160(5 Pt 1): 1743-9, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10556150

ABSTRACT

The effect of 3-aminobenzamide, an inhibitor of poly (ADP-ribose) synthetase activity, was evaluated in a rat model of laryngeal injury induced by endotracheal intubation for 1 h. At 1 h after extubation, the laryngeal damage was characterized by areas of mucosal necrosis, submucosal edema, swelling of subglottic glands, and submucosal infiltration of inflammatory cells. Activity of myeloperoxidase, a marker of neutrophil infiltration, was also markedly increased into the damaged tissue. Immunohistochemistry for nitrotyrosine, an index of nitrosative stress, showed an intense staining in the inflamed larynx. Treatment with 3-aminobenzamide (10 mg/kg intraperitoneally) significantly reduced the appearance of mucosal damage and was associated with a significant reduction of tissue myeloperoxidase activity and nitrotyrosine immunoreactivity in the larynx. The results of this study suggest that poly (ADP-ribose) synthetase may play a role in the inflammatory process after laryngeal intubation and extubation, and administration of 3-aminobenzamide may be a beneficial therapeutic approach.


Subject(s)
Benzamides/pharmacology , Enzyme Inhibitors/pharmacology , Intubation, Intratracheal/adverse effects , Larynx/injuries , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Immunohistochemistry , Laryngeal Mucosa/chemistry , Laryngeal Mucosa/pathology , Larynx/pathology , Male , Neutrophils/pathology , Peroxidase/analysis , Rats , Rats, Wistar , Tyrosine/analogs & derivatives , Tyrosine/analysis , Wounds and Injuries/etiology , Wounds and Injuries/pathology
12.
Crit Care Med ; 27(9): 1701-7, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10507586

ABSTRACT

OBJECTIVE: Nitric oxide (NO), produced by the inducible isoform of NO synthase (NOS) in circulatory shock exerts cytotoxic and vasodilator effects. Part of these effects are mediated by formation of peroxynitrite, a toxic oxidant produced by the rapid reaction of NO and superoxide. Other parts of the vascular actions of NO in shock are thought to be mediated by the action of NO on the soluble guanylyl cyclase (GC) in the smooth muscle and subsequent decrease in the intracellular calcium levels. Using 1H-(1,2,4)oxadiazolo(4,3-alpha)quinoxalin-1 -one (ODQ), a potent inhibitor of GC, we studied the role of GC activation in the NO- and peroxynitrite-related vascular alterations. DESIGN: In vitro: Controlled experiment using cultured rat aortic smooth muscle cells. In vivo: Prospective, randomized, controlled animal study. SETTING: Experimental laboratory. SUBJECTS: Male Wistar rats and male Swiss mice. INTERVENTIONS: In vitro: a) Stimulation of rat aortic smooth muscle cells with bacterial lipopolysaccharide (LPS) and gamma-interferon, measurement of the production of nitrite and nitrate (breakdown products of NO), and suppression of mitochondrial respiration for 24 to 48 hrs, in the presence or absence of ODQ; and b) in norepinephrine-precontracted endothelium-denuded thoracic aortic rings, exposure to LPS (10 ng/mL) in the presence or absence of ODQ. In vivo: Rats treated in vivo with LPS (10 mg/kg iv for 3 hrs) and mice challenged with 60 mg/kg LPS ip, in the presence or absence of ODQ. MEASUREMENTS AND MAIN RESULTS: Stimulation of rat aortic smooth muscle cells with bacterial LPS and gamma-interferon induced the production of nitrite and nitrate (breakdown products of NO) and suppression of mitochondrial respiration for 24 to 48 hrs. The amount of NO produced was slightly enhanced with ODQ (10-100 EM), whereas the suppression of mitochondrial respiration was not affected by ODQ (1-100 microM). ODQ did not affect the degree of suppression of mitochondrial respiration in response to NO donor agents or to peroxynitrite. Exposure to LPS (10 ng/mL) for 6 hrs caused a time-dependent relaxation of norepinephrine-precontracted endothelium-denuded thoracic aortic rings. This response was caused by the expression of inducible NOS and could be blocked by pharmacologic inhibitors of NOS such as N(G)-methylL-arginine. ODQ (1 microM) prevented the LPS-induced loss of vascular tone in this experimental system. Similar to the in vitro responses, there was a significant suppression of the norepinephrine-induced contractions in ex vivo experiments, in which rings were taken from animals treated in vivo with LPS (10 mg/kg for 3 hrs). ODQ treatment in vitro (1 microM) caused a complete restoration of the contractile responses. In mice challenged with 60 mg/kg LPS ip, ODQ (20 mg/kg), given either as a pretreatment or as a 4-hr posttreatment, improved survival at 24-144 hrs. CONCLUSION: These studies indicate that GC activation does not contribute to NO- or peroxynitrite-induced cytotoxicity but does contribute to the vascular hyporeactivity induced by endotoxin in vitro and in vivo. GC inhibition alone is sufficient to influence survival in a murine model of severe sepsis.


Subject(s)
Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Muscle, Smooth, Vascular/enzymology , Nitrates/metabolism , Nitric Oxide/metabolism , Oxadiazoles/pharmacology , Oxidants/metabolism , Quinoxalines/pharmacology , Shock, Septic/enzymology , Animals , Aorta/enzymology , Cell Respiration/drug effects , Cells, Cultured , In Vitro Techniques , Interferon-gamma/pharmacology , Lipopolysaccharides , Male , Mice , Mitochondria/metabolism , Muscle, Smooth, Vascular/drug effects , Nitrites/metabolism , Rats , Rats, Wistar , Survival Analysis , Vasodilation/drug effects
13.
Crit Care Med ; 27(8): 1517-23, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10470759

ABSTRACT

BACKGROUND AND METHODS: In the present study, we tested the hypothesis that peroxynitrite and subsequent activation of the nuclear enzyme poly(ADP-ribose) synthetase (PARS) play a role in the pathogenesis of multiple organ failure induced by peritoneal injection of zymosan in the rat. Animals were randomly divided into six groups (ten rats for each group). The first group was treated with ip administration of saline solution (0.9% NaCl) and served as the sham group. The second group was treated with ip administration of zymosan (500 mg/kg suspended in saline solution). In the third and fourth groups, rats received ip administration of 3-aminobenzamide (10 mg/kg) 1 and 6 hrs after zymosan or saline administration, respectively. In the fifth and sixth groups, rats received ip administration of nicotinamide (50 mg/kg) 1 and 6 hrs after zymosan or saline administration, respectively. After zymosan or saline injection, animals were monitored for 72 hrs to evaluate systemic toxicity (conjunctivitis, ruffled fur, diarrhea, and lethargy), loss of body weight, and mortality. RESULTS: A severe inflammatory response, characterized by peritoneal exudation, high plasma and peritoneal levels of nitrate/nitrite (the breakdown products of nitric oxide), and leukocyte infiltration into peritoneal exudate, was induced by zymosan administration. This inflammatory process coincided with the damage of lung, small intestine, and liver as assessed by histologic examination and by an increase of myeloperoxidase activity, which is indicative of neutrophil infiltration. Zymosan-treated rats showed signs of systemic illness, significant loss of body weight, and high mortality rates. Peritoneal administration of zymosan in the rat also induced a significant increase in the plasma levels of peroxynitrite as measured by the oxidation of the fluorescent dihydrorhodamine 123. Immunohistochemical examination demonstrated a marked increase in the immunoreactivity to nitrotyrosine, a specific "footprint" of peroxynitrite, in the lung of zymosan-shocked rats. In vivo treatment with ip administration of 3-aminobenzamide (10 mg/kg, 1 and 6 hrs after zymosan injection) or nicotinamide (50 mg/kg, 1 and 6 hrs after zymosan injection) significantly decreased mortality, inhibited the development of peritonitis, and reduced peroxynitrite formation. In addition, PARS inhibitors were effective in preventing the development of organ failure because tissue injury and neutrophil infiltration, by myeloperoxidase evaluation, were reduced in the lung, small intestine, and liver. CONCLUSIONS: In conclusion, the major findings of our study are that peroxynitrite and the consequent PARS activation exert a role in the development of multiple organ failure and that PARS inhibition is an effective anti-inflammatory therapeutic tool.


Subject(s)
Benzamides/therapeutic use , Multiple Organ Failure/drug therapy , Multiple Organ Failure/enzymology , Niacinamide/therapeutic use , Peritonitis/complications , Poly(ADP-ribose) Polymerase Inhibitors , Zymosan , Animals , Benzamides/immunology , Body Weight/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Male , Multiple Organ Failure/etiology , Multiple Organ Failure/immunology , Niacinamide/immunology , Nitrates/immunology , Peritonitis/chemically induced , Peritonitis/mortality , Peritonitis/pathology , Random Allocation , Rats , Rats, Sprague-Dawley , Time Factors
14.
Life Sci ; 65(9): 957-64, 1999.
Article in English | MEDLINE | ID: mdl-10465355

ABSTRACT

The aim of the present study was to investigate the role of poly (ADP-ribose) synthetase (PARS) in a model of acute local inflammation (zymosan-activated plasma (ZAP)-induced paw edema), in which the oxyradicals, nitric oxide and peroxynitrite, are known to play a crucial role. Injection of zymosan-activated plasma (ZAP) into the rat paw induced edema formation. The maximal increase in paw volume was observed at three hours after administration (maximal in paw volume: 1.29+/-0.09 ml). At this time point, there was a marked increase in neutrophil infiltration in the paw, as measured by an increase in myeloperoxidase (MPO) activity in the paw tissue (260+/-25 mU/100 mg wet tissue). However, ZAP-induced paw edema was significantly reduced in a dose-dependent manner by treatment with 3-aminobenzamide (3-AB) or nicotinamide (NIC), two inhibitors of PARS, at 1, 2, 3, 4 hours after ZAP injection. PARS inhibition also caused a significant reduction of MPO activity. The paw tissues were also examined immunohistochemically for the presence of nitrotyrosine (a footprint for peroxynitrite formation). At 3 h following ZAP injection, staining for nitrotyrosine were also found to be localised within discrete cells in the inflamed paw tissue. Treatment with PARS inhibitor prevented the appearance of nitrotyrosine in the tissues. Our results suggest that in paw edema induced by ZAP, inhibition of PARS exert potent anti-inflammatory effects.


Subject(s)
Benzamides/pharmacology , Inflammation/enzymology , Niacinamide/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Complement Activation , Dose-Response Relationship, Drug , Edema/enzymology , Edema/immunology , Edema/metabolism , Enzyme Inhibitors/pharmacology , Free Radicals/metabolism , Immunohistochemistry , Inflammation/immunology , Inflammation/metabolism , Male , Nitrates/metabolism , Peroxidase/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Rats , Rats, Wistar , Tyrosine/analogs & derivatives , Tyrosine/analysis , Zymosan/pharmacology
15.
Eur J Pharmacol ; 372(2): 187-97, 1999 May 14.
Article in English | MEDLINE | ID: mdl-10395099

ABSTRACT

In the present study we investigated the protective role of endogenous glutathione, a known free radical scavenger, in rats subjected to carrageenan-induced pleurisy. In vivo depletion of endogenous glutathione pools with L-buthionine-(S,R)-sulfoximine (BSO, 1 g/kg for 24 h, intraperitoneally) enhances the carrageenan-induced degree of pleural exudation and polymorphonuclear leukocyte migration in rats subjected to carrageenan-induced pleurisy. Lung myeloperoxidase activity and lipid peroxidation were significantly increased in BSO pretreated rats. However, the inducible nitric oxide (NO) synthase in lung samples was unaffected by BSO pretreatment. Immunohistochemical analysis for nitrotyrosine revealed a positive staining in lungs from carrageenan-treated rats, which was massively enhanced by BSO pretreatment. Furthermore, in vivo BSO pretreatment significantly increased peroxynitrite formation as measured by the oxidation of the fluorescent dye dihydrorhodamine 123, enhanced the appearance of DNA damage, the decrease in mitochondrial respiration and partially decreased the cellular level of NAD+ in ex vivo macrophages harvested from the pleural cavity of rats subjected to carrageenan-induced pleurisy. In vivo treatment with exogenous glutathione (50 mg/kg i.p.) significantly reverts the effects of BSO and exerts anti-inflammatory effects. Thus, endogenous glutathione plays an important protective role against carrageenan-induced local inflammation.


Subject(s)
Glutathione/physiology , Pleurisy/metabolism , Animals , Carrageenan , Cells, Cultured , Disease Models, Animal , Free Radical Scavengers/metabolism , Glutathione/deficiency , Macrophages/metabolism , Macrophages/physiology , Nitrates/metabolism , Nitric Oxide/metabolism , Oxidants/metabolism , Pleurisy/chemically induced , Protective Agents , Rats , Tyrosine/analogs & derivatives , Tyrosine/metabolism
16.
Gut ; 45(2): 199-209, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10403731

ABSTRACT

BACKGROUND: Oxidative and nitrosative stress have been implicated in the pathogenesis of inflammatory bowel diseases. AIMS: To study the role of nitric oxide (NO) derived from inducible NO synthase (iNOS) in an experimental model of murine enterocolitis. METHODS: Trinitrobenzene sulphonic acid (TNBS) was instilled per rectum to induce a lethal colitis in iNOS deficient mice and in wild type controls. The distal colon was evaluated for histological evidence of inflammation, iNOS expression and activity, tyrosine nitration and malondialdehyde formation (as indexes of nitrosative and oxidative stress), myeloperoxidase activity (as index of neutrophil infiltration), and tissue localisation of intercellular adhesion molecule 1 (ICAM-1). RESULTS: TNBS administration induced a high mortality and weight loss associated with a severe colonic mucosal erosion and ulceration, increased myeloperoxidase activity, increased concentrations of malondialdehyde, and an intense staining for nitrotyrosine and ICAM-1 in wild type mice. Genetic ablation of iNOS gene conferred to mice a significant resistance to TNBS induced lethality and colonic damage, and notably reduced nitrotyrosine formation and concentrations of malondialdehyde; it did not, however, affect neutrophil infiltration and intestinal ICAM-1 expression in the injured tissue. CONCLUSION: Data show that activation of iNOS is required for nitrosative and oxidative damage in experimental colitis.


Subject(s)
Enterocolitis/enzymology , Nitric Oxide Synthase/metabolism , Nitric Oxide/physiology , Oxidative Stress , Animals , Enterocolitis/chemically induced , Enzyme Activation , Female , Immunohistochemistry , Intercellular Adhesion Molecule-1/metabolism , Male , Malondialdehyde/metabolism , Mice , Nitric Oxide Synthase Type II , Trinitrobenzenesulfonic Acid
17.
Biochem Pharmacol ; 58(1): 171-6, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10403531

ABSTRACT

In the present study we investigated the therapeutic efficacy of Mn(III)tetrakis (4-benzoic acid) porphyrin (MnTBAP), a superoxide dismutase mimetic which possesses peroxynitrite scavenging effects, in rats subjected to carrageenan-induced paw oedema. Local administration of MnTBAP (5, 25, and 50 microg/paw) significantly and dose dependently reduced carrageenan-induced paw oedema at all time points. MnTBAP also caused a significant dose-dependent reduction in paw myeloperoxidase activity and lipid peroxidation, as well as preventing histological injury. Immunohistochemical analysis for nitrotyrosine revealed a positive staining in paw from carrageenan-treated rats. No positive nitrotyrosine staining was found in the paws of the carrageenan-treated rats that received MnTBAP. Our study demonstrates that MnTBAP exerts protective effects in carrageenan-induced paw oedema. Part of these anti-inflammatory effects may be related to: 1) reduction of superoxide formation due to the superoxide dismutase-like activity of the compound; and 2) scavenging of peroxynitrite.


Subject(s)
Edema/prevention & control , Free Radical Scavengers/therapeutic use , Metalloporphyrins/therapeutic use , Protective Agents/therapeutic use , Animals , Carrageenan , Dose-Response Relationship, Drug , Edema/chemically induced , Edema/pathology , Male , Molecular Mimicry , Peroxidase/metabolism , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism
18.
Gastroenterology ; 116(2): 335-45, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9922314

ABSTRACT

BACKGROUND & AIMS: Inflammatory bowel disease is characterized by oxidative and nitrosative stress, leukocyte infiltration, and up-regulation of intercellular adhesion molecule 1 (ICAM-1) expression in the colon. Recent data show that oxidative and nitrosative stress in isolated enterocytes produces DNA single-strand breaks that activate the nuclear enzyme poly(ADP-ribose) synthetase (PARS), resulting in depletion of intracellular energetics and increased paracellular permeability. The aim of the present study was to examine the in vivo relevance of this injury pathway. METHODS: Colitis was induced by rectal instillation of trinitrobenzenesulfonic acid (TNBS) in mice with a genetic deficiency of PARS (PARS-/-) and in wild-type littermates. RESULTS: In wild-type mice, TNBS treatment resulted in colonic erosion and ulceration that was maintained up to 7 days. Neutrophil infiltration (indicated by myeloperoxidase activity in the mucosa) was associated with up-regulation of ICAM-1 and high levels of malondialdehyde and nitrotyrosine. TNBS-treated PARS-/- mice experienced a similar colonic injury that was, however, completely resolved by 6 days. Resolution of the damage was associated with absence of ICAM-1 up-regulation, reduction of neutrophil infiltration, lipid peroxidation, and nitrosative damage. CONCLUSIONS: These data show that PARS plays a critical role in colonic inflammation possibly by regulating ICAM-1 expression, neutrophil recruitment, and the subsequent oxidant generation.


Subject(s)
Colitis/metabolism , Colitis/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Neutrophils , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Colitis/chemically induced , Colitis/enzymology , Disease Models, Animal , Immunohistochemistry , Intercellular Adhesion Molecule-1/metabolism , Intestinal Mucosa/enzymology , Malondialdehyde/metabolism , Mice , Oxidative Stress , Permeability , Peroxidase/metabolism , Severity of Illness Index , Time Factors , Trinitrobenzenesulfonic Acid , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Up-Regulation
19.
Free Radic Biol Med ; 26(1-2): 25-33, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9890637

ABSTRACT

Peroxynitrite, a potent cytotoxic oxidant formed by the reaction of NO with superoxide anion, has been proposed to have major pathogenetic role in inflammatory process. Here we have investigated the therapeutic efficacy of Mn(III)tetrakis (4-benzoic acid) porphyrin (MnTBAP), a novel superoxide dismutase mimetic that possesses peroxynitrite scavenging effect, in rats subjected to carrageenan-induced pleurisy. In vivo treatment with MnTBAP (3 and 10 mg/kg 5 min before carrageenan) prevented in a dose-dependent manner the carrageenan-induced the degree of pleural exudation, polymorphonuclear migration in rats subjected to carrageenan-induced pleurisy. Lung myeloperoxidase (MPO) activity and histological organ injury was significantly reduced by MnTBAP. However, MnTBAP did not inhibit the inducible NO synthase in lung samples. Immunohistochemical analysis for nitrotyrosine, a footprint of peroxynitrite, revealed a positive staining in lungs from carrageenan-treated rats. No positive nitrotyrosine staining was found in the lungs of the carrageenan-treated rats that received MnTBAP (10 mg/kg) treatment. In addition, in vivo MnTBAP treatment significantly reduced in a dose-dependent manner peroxynitrite formation as measured by the oxidation of the fluorescent dye dihydrorhodamine 123, prevented the appearance of DNA damage, the decrease in mitochondrial respiration and partially restored the cellular level of NAD+ in ex vivo macrophages harvested from the pleural cavity of rats subjected to carrageenan-induced pleurisy. Our study demonstrates that the MnTBAP exerts multiple protective effects in carrageenan-induced pleurisy. We suggest peroxynitrite produced during the inflammatory process trigger DNA strand breakage and subsequent cellular dysfunction. Part of these anti-inflammatory effects may be related to: (1) reduction of superoxide formation due to the superoxide dismutase-like activity of the compound and (2) scavenging of peroxynitrite.


Subject(s)
Free Radical Scavengers/pharmacology , Metalloporphyrins/pharmacology , Pleurisy/drug therapy , Superoxide Dismutase/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carrageenan/toxicity , DNA Damage , Energy Metabolism/drug effects , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Neutrophils/drug effects , Neutrophils/pathology , Nitrates/metabolism , Nitric Oxide/biosynthesis , Pleurisy/chemically induced , Pleurisy/metabolism , Rats , Rats, Sprague-Dawley , Superoxides/metabolism
20.
J Pharmacol Exp Ther ; 287(3): 1048-55, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9864291

ABSTRACT

The effect of mercaptoethylguanidine (MEG), a selective inhibitor of the inducible nitric oxide synthase and peroxynitrite scavenger, was evaluated in a rat model of colonic injury. A single intracolonic administration of trinitrobenzene sulfonic acid (TNBS, 20 mg/kg) dissolved in ethanol induced a severe colitis in male rats. Rats experienced bloody diarrhea and a significant loss of body weight. At 4 days after TNBS administration, the colon damage was characterized by areas of mucosal necrosis. Activity of myeloperoxidase, a marker of neutrophil infiltration, and levels of the 6-keto-prostaglandin F1alpha, were also markedly increased, whereas colonic ATP levels were reduced into the damaged tissue. Immunohistochemistry for the inducible nitric oxide synthase and nitrotyrosine, an index of nitrosative stress, showed an intense staining in the inflamed colon. Treatment with MEG (10 mg/kg i.v. b. i.d.) significantly reduced the appearance of diarrhea and the loss of body weight. This was associated with a remarkable amelioration of the disruption of the colonic architecture and suppression of the energetic failure, as well as a significant reduction of colonic myeloperoxidase activity and 6-keto-prostaglandin F1alpha levels. MEG also reduced the appearance of iNOS and nitrotyrosine immunoreactivity in the colon. The results of this study suggested that administration of MEG may be beneficial for the treatment of inflammatory bowel diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Colitis/prevention & control , Free Radical Scavengers/pharmacology , Guanidines/pharmacology , Nitrates/analysis , Nitric Oxide Synthase/antagonists & inhibitors , Animals , Body Weight/drug effects , Colitis/chemically induced , Enzyme Inhibitors/pharmacology , Immunohistochemistry , Male , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase Type II , Rats , Rats, Wistar , Trinitrobenzenesulfonic Acid , Tyrosine/analogs & derivatives , Tyrosine/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...