Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Osteoarthritis Cartilage ; 21(1): 151-6, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23025928

ABSTRACT

OBJECTIVES: To characterize the utility of nitrotyrosine (NT) as a biomarker for arthritis and joint injury. DESIGN: Synovial fluid, plasma, and urine from patients diagnosed with osteoarthritis (OA), rheumatoid arthritis (RA), anterior cruciate ligament (ACL) injury, meniscus injury and pseudogout, and knee-healthy volunteers were analyzed for concentrations of NT, nitrate and nitrite (NO(x)), matrix metalloproteinase (MMP)-3, MMP-1, MMP-9, more than 40 chemokines and cytokines. RESULTS: In OA, plasma and synovial fluid NT were increased versus healthy volunteers. Synovial fluid to plasma NT ratios were elevated in OA patients. Synovial fluid from patients with ACL and meniscus injury and pseudogout had increased levels of NT (P < 0.001). In these samples, NT levels significantly correlated with ARGS-aggrecan neoepitope generated by aggrecanase cleavage of aggrecan (P ≤ 0.001), cross-linked C-telopeptides of type II collagen (P < 0.001), MMP-1 (P = 0.008), and MMP-3 (P ≤ 0.001). In RA, plasma NT decreased following 6 months of anti-tumor necrosis factor (TNF) treatment. For every 1.1% change in log(10) NT, there was a 1.0% change in the log(10) disease activity scores (DAS28-3 CRP). Both predicted and observed DAS28-3 CRP showed a robust linear relationship with NT. RA plasma NT positively correlated with CRP, MMP-3 and interferon γ-induced protein 10. CONCLUSIONS: NT may serve as a useful biomarker for arthritis and joint injury. In RA, NT is highly correlated with several biomarkers and clinical correlates of disease activity and responds to anti-TNF therapy.


Subject(s)
Anterior Cruciate Ligament/metabolism , Arthritis, Rheumatoid/metabolism , Chondrocalcinosis/metabolism , Menisci, Tibial/metabolism , Osteoarthritis, Knee/metabolism , Anterior Cruciate Ligament Injuries , Case-Control Studies , Chemokines/metabolism , Cytokines/metabolism , Female , Humans , Male , Matrix Metalloproteinases, Secreted/metabolism , Nitrates/metabolism , Synovial Fluid/metabolism , Tibial Meniscus Injuries , Tyrosine/analogs & derivatives , Tyrosine/metabolism
2.
Proc Natl Acad Sci U S A ; 97(17): 9753-8, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10944234

ABSTRACT

A major feature of septic shock is the development of a vascular crisis characterized by nonresponsiveness to sympathetic vasoconstrictor agents and the subsequent irreversible fall in blood pressure. In addition, sepsis, like other inflammatory conditions, results in a large increase in the production of free radicals, including superoxide anions (O(2)) within the body. Here we show that O(2) reacts with catecholamines deactivating them in vitro. Moreover, this deactivation would appear to account for the hyporeactivity to exogenous catecholamines observed in sepsis, because administration of a superoxide dismutase (SOD) mimetic to a rat model of septic shock to remove excess O(2) restored the vasopressor responses to norepinephrine. This treatment with the SOD mimetic also reversed the hypotension in these animals; suggesting that deactivation of endogenous norepinephrine by O(2) contributes significantly to this aspect of the vascular crisis. Indeed, the plasma concentrations of both norepinephrine and epinephrine in septic rats treated with the SOD mimetic were significantly higher than in untreated rats. Interestingly, the plasma concentrations for norepinephrine and epinephrine were inversely related to the plasma concentrations of adrenochromes, the product of the autoxidation of catecholamines initiated by O(2). We propose, therefore, that the use of a SOD mimetic represents a new paradigm for the treatment of septic shock. By removing O(2), exogenous and endogenous catecholamines are protected from autoxidation. As a result, both hyporeactivity and hypotension are reversed, generation of potentially toxic adrenochromes is reduced, and survival rate is improved.


Subject(s)
Catecholamines/antagonists & inhibitors , Catecholamines/metabolism , Shock, Septic/metabolism , Shock, Septic/pathology , Superoxides/metabolism , Superoxides/pharmacology , Adrenochrome/blood , Animals , Blood Pressure/drug effects , Catecholamines/blood , Catecholamines/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Epinephrine/antagonists & inhibitors , Epinephrine/blood , Epinephrine/metabolism , Hypotension/blood , Hypotension/chemically induced , Hypotension/metabolism , Hypotension/pathology , Kinetics , Lipopolysaccharides/pharmacology , Male , Manganese , Norepinephrine/antagonists & inhibitors , Norepinephrine/blood , Norepinephrine/metabolism , Norepinephrine/pharmacology , Organometallic Compounds/pharmacology , Rats , Rats, Sprague-Dawley , Shock, Septic/blood , Shock, Septic/chemically induced , Superoxide Dismutase/metabolism , Superoxides/antagonists & inhibitors , Vasoconstriction/drug effects , Vasoconstrictor Agents/antagonists & inhibitors , Vasoconstrictor Agents/blood , Vasoconstrictor Agents/metabolism , Vasoconstrictor Agents/pharmacology
3.
FASEB J ; 14(9): 1061-72, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10834927

ABSTRACT

The aim of the present study was to investigate the protective effect of the peroxynitrite decomposition catalyst 5,10,15, 20-tetrakis(2,4,6-trimethyl-3,5-disulfonatophenyl)-porphyrinato iron (III) (FeTMPS) in a model of splanchnic artery occlusion shock (SAO). SAO shock was induced in rats by clamping both the superior mesenteric artery and the celiac trunk for 45 min, followed by release of the clamp (reperfusion). At 60 min after reperfusion, animals were killed for histological examination and biochemical studies. There was a marked increase in the oxidation of dihydrorhodamine 123 to rhodamine (a marker of peroxynitrite-induced oxidative processes) in the plasma of the SAO-shocked rats after reperfusion, but not during ischemia alone. Immunohistochemical examination demonstrated a marked increase in the immunoreactivity to nitrotyrosine, an index of nitrogen species such as peroxynitrite, in the necrotic ileum in shocked rats. SAO-shocked rats developed a significant increase of tissue myeloperoxidase and malonaldehyde activity, and marked histological injury to the distal ileum. SAO shock was also associated with a significant mortality (0% survival at 2 h after reperfusion). Reperfused ileum tissue sections from SAO-shocked rats showed positive staining for P-selectin localized mainly in the vascular endothelial cells. Ileum tissue sections obtained from SAO-shocked rats and stained with antibody to ICAM-1 showed a diffuse staining. Administration of FeTMPS significantly reduced ischemia/reperfusion injury in the bowel, and reduced lipid and the production of peroxynitrite during reperfusion. Treatment with PN catalyst also markedly reduced the intensity and degree of P-selectin and ICAM-1 staining in tissue sections from SAO-shocked rats and improved survival. Our results clearly demonstrate that peroxynitrite decomposition catalysts exert a protective effect in SAO and that this effect may be due to inhibition of the expression of adhesion molecules and the tissue damage associated with peroxynitrite-related pathways.


Subject(s)
Ferric Compounds/therapeutic use , Ileum/blood supply , Ileum/pathology , Metalloporphyrins/therapeutic use , Nitrates/metabolism , Reperfusion Injury/drug therapy , Splanchnic Circulation/physiology , Animals , Blood Pressure/drug effects , Catalysis/drug effects , Celiac Artery/physiology , Disease Models, Animal , Endothelium, Vascular/chemistry , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Ferric Compounds/pharmacology , Ileum/drug effects , Ileum/metabolism , Intercellular Adhesion Molecule-1/analysis , Leukocyte Count/drug effects , Lipid Peroxidation/drug effects , Male , Malondialdehyde/blood , Malondialdehyde/metabolism , Metalloporphyrins/pharmacology , Nitrates/blood , Nitric Oxide/blood , Nitric Oxide/metabolism , Nitrites/blood , Nitrites/metabolism , Oxidative Stress/drug effects , P-Selectin/analysis , Peroxidase/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/mortality , Reperfusion Injury/pathology , Rhodamines/blood , Rhodamines/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
4.
J Neuroimmunol ; 107(1): 21-8, 2000 Jul 10.
Article in English | MEDLINE | ID: mdl-10808047

ABSTRACT

Peroxynitrite (PN), the product of nitric oxide (NO) reacted with superoxide, is generated at sites of inflammation. Nitrotyrosine (NT), a marker of PN formation, is abundant in lesions of acute experimental autoimmune encephalomyelitis (EAE), and in active multiple sclerosis (MS) plaques. To determine whether PN plays a role in EAE pathogenesis, mice induced to develop EAE were treated with a catalyst specific for the decomposition of PN. Because this catalyst has no effect upon NO, using it allowed differentiation of PN-mediated effects from NO-mediated effects. Mice receiving the PN decomposition catalyst displayed less severe clinical disease, and less inflammation and demyelination than control mice. Encephalitogenic T cells could be recovered from catalyst-treated mice, indicating that the PN decomposition catalyst blocked the pathogenic action of PN at the effector stage of EAE, but was not directly toxic to encephalitogenic T cells. PN plays an important role distinct from that of NO in the pathogenesis of EAE, a major model for MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/prevention & control , Ferric Compounds/pharmacology , Metalloporphyrins/pharmacology , Nitrates/metabolism , Animals , Catalysis , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Immunohistochemistry , Lymphocyte Transfusion , Lymphocytes/immunology , Mice , Mice, Inbred Strains , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Tyrosine/analogs & derivatives , Tyrosine/metabolism
5.
Science ; 286(5438): 304-6, 1999 Oct 08.
Article in English | MEDLINE | ID: mdl-10514375

ABSTRACT

Many human diseases are associated with the overproduction of oxygen free radicals that inflict cell damage. A manganese(II) complex with a bis(cyclohexylpyridine)-substituted macrocyclic ligand (M40403) was designed to be a functional mimic of the superoxide dismutase (SOD) enzymes that normally remove these radicals. M40403 had high catalytic SOD activity and was chemically and biologically stable in vivo. Injection of M40403 into rat models of inflammation and ischemia-reperfusion injury protected the animals against tissue damage. Such mimics may result in better clinical therapies for diseases mediated by superoxide radicals.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Inflammation/drug therapy , Organometallic Compounds/toxicity , Reperfusion Injury/drug therapy , Superoxide Dismutase , Superoxides/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Cytoprotection , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Drug Design , Drug Stability , Interleukin-1/metabolism , L-Lactate Dehydrogenase/metabolism , Male , Manganese , Molecular Mimicry , Neutrophils/drug effects , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Organometallic Compounds/metabolism , Rats , Rats, Sprague-Dawley , Splanchnic Circulation , Superoxide Dismutase/metabolism , Time Factors , Tumor Necrosis Factor-alpha/metabolism
6.
Br J Pharmacol ; 127(3): 685-92, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10401559

ABSTRACT

1. The relative contributions of superoxide anion (O2-) and peroxynitrite (PN) were evaluated in the pathogenesis of intestinal microvascular damage caused by the intravenous injection of E. coli lipopolysaccharide (LPS) in rats. The superoxide dismutase mimetic (SODm) SC-55858 and the active peroxynitrite decomposition catalysts 5,10,15,20-tetrakis(2,4,6-trimethyl-3,5-disulphonatophenyl)-por phyrinato iron (III) and 5,10,15,20-tetrakis(N-methyl-4'-pyridyl)-porphyrinato iron (III) (FeTMPS, FeTMPyP respectively) were used to assess the roles of O2- and PN respectively. 2. The intravenous injection of LPS elicited an inflammatory response that was characterized by a time-dependent infiltration of neutrophils, lipid peroxidation, microvascular leakage (indicative of microvascular damage), and epithelial cell injury in both the duodenum and jejunum. 3. Administration of the SODm SC-55858, FeTMPS or FeTMPyP at 3 h post LPS reduced the subsequent increase in microvascular leakage, lipid peroxidation and epithelial cell injury. Inactive peroxynitrite decomposition catalysts exhibited no protective effects. Only, SC-55858 inhibited neutrophil infiltration. 4. Our results suggest that O2 and peroxynitrite play a significant role in the pathogenesis of duodenal and intestinal injury during endotoxaemia and that their remoyal by SODm and peroxynitrite decomposition catalysts offers a novel approach to the treatment of septic shock or clinical conditions of gastrointestinal inflammation. Furthermore, the remarkable protection of the intestinal epithelium by these agents suggests their use during chemo- and radiation therapy, cancer treatments characterized by gastrointestinal damage. Potential mechanisms through which these radicals evoke damage are discussed.


Subject(s)
Free Radical Scavengers/pharmacology , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Lipopolysaccharides/toxicity , Nitrates/toxicity , Organometallic Compounds/pharmacology , Oxidants/toxicity , Superoxide Dismutase/pharmacology , Animals , Catalysis , Colchicine/pharmacology , Ferric Compounds/pharmacology , Free Radical Scavengers/metabolism , Intestinal Mucosa/blood supply , Intestinal Mucosa/metabolism , Lipid Peroxidation/drug effects , Male , Metalloporphyrins/pharmacology , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/pathology , Nitrates/metabolism , Organometallic Compounds/metabolism , Oxidants/metabolism , Porphyrins/pharmacology , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism , Superoxides/metabolism
7.
Ann Thorac Surg ; 67(3): 716-22, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10215216

ABSTRACT

BACKGROUND: We have demonstrated that inhibition of inducible nitric oxide synthase (NOS) ameliorated acute cardiac allograft rejection. This study determined the time course and cellular localization of inducible NOS expression during the histologic progression of unmodified acute rat cardiac allograft rejection. METHODS: Tissue from syngeneic (ACI to ACI) and allogeneic (Lewis to ACI) transplants were harvested on postoperative days 3 through 10 and analyzed for inducible NOS mRNA expression (ribonuclease protection assay), inducible NOS enzyme activity (conversion of L-[3H]arginine to nitric oxide and L-[3H]citrulline), and nitric oxide production (serum nitrite/nitrate levels). Inducible NOS mRNA and protein expression were localized using in situ hybridization and immunohistochemistry. RESULTS: Inducible NOS mRNA and enzyme activity were expressed in allografts during mild, moderate, and severe acute rejection (postoperative days 4 through 10), but were not detected in normals, isografts, or allografts before histologic changes of mild acute rejection (postoperative day 3). Inducible NOS expression resulted in increased serum nitrite/nitrate levels during mild and moderate rejection (postoperative days 4 through 6). Inducible NOS mRNA and protein expression localized to infiltrating mononuclear inflammatory cells in allograft tissue sections during all stages of rejection but were not detected in allograft parenchymal cells or in normals or isografts. CONCLUSIONS: Inducible NOS expression and increased nitric oxide production occurred during the early stages of acute rejection, persisted throughout the unmodified rejection process, and localized to infiltrating inflammatory cells but not allograft parenchymal cells during all stages of acute rejection.


Subject(s)
Graft Rejection/enzymology , Heart Transplantation , Myocardium/enzymology , Nitric Oxide Synthase/metabolism , Acute Disease , Animals , Graft Rejection/metabolism , Immunohistochemistry , In Situ Hybridization , Male , Myocardium/cytology , Myocardium/metabolism , Nitrates/blood , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitrites/blood , RNA, Messenger/analysis , Rats , Rats, Inbred ACI , Rats, Inbred Lew , Transplantation, Isogeneic
8.
J Neuroimmunol ; 88(1-2): 45-56, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9688323

ABSTRACT

Peroxynitrite, generated by the reaction of nitric oxide (NO) with superoxide at sites of inflammation, is a strong oxidant capable of damaging tissues and cells. Detection of nitrotyrosine (NT) at inflammatory sites serves as a biochemical marker for peroxynitrite-mediated damage. In this study, NT was detected immunohistochemically within autopsied CNS tissues from six of nine multiple sclerosis (MS) patients, and in most of the MS sections displaying inflammation. Nitrite and nitrate, the stable oxidation products of NO and peroxynitrite, respectively, were measured in cerebrospinal fluid samples obtained from MS patients and controls. Levels of nitrate were elevated significantly during clinical relapses of MS. These data suggest that peroxynitrite formation is a major consequence of NO produced in MS-affected CNS and implicate a role for this powerful oxidant in the pathogenesis of MS.


Subject(s)
Central Nervous System/metabolism , Multiple Sclerosis/metabolism , Nitrates/metabolism , Humans , Immunohistochemistry , Inflammation/cerebrospinal fluid , Multiple Sclerosis/cerebrospinal fluid , Nervous System Diseases/cerebrospinal fluid , Nitrates/cerebrospinal fluid , Nitrites/cerebrospinal fluid , Recurrence , Tyrosine/analogs & derivatives , Tyrosine/metabolism
9.
J Biol Chem ; 273(25): 15646-53, 1998 Jun 19.
Article in English | MEDLINE | ID: mdl-9624158

ABSTRACT

The formation of the powerful oxidant peroxynitrite (PN) from the reaction of superoxide anion with nitric oxide has been shown to be a kinetically favored reaction contributing to cellular injury and death at sites of tissue inflammation. The PN molecule is highly reactive causing lipid peroxidation as well as nitration of both free and protein-bound tyrosine. We present evidence for the pharmacological manipulation of PN with decomposition catalysts capable of converting it to nitrate. In target cells challenged with exogenously added synthetic PN, a series of metalloporphyrin catalysts (5,10,15,20-tetrakis(2,4,6-trimethyl-3, 3-disulfonatophenyl)porphyrinato iron (III) (FeTMPS); 5,10,15, 20-tetrakis(4-sulfonatophenyl)porphyrinato iron (III) (FeTPPS); 5,10, 15,20-tetrakis(N-methyl-4'-pyridyl)porphyrinato iron (III) (FeTMPyP)) provided protection against PN-mediated injury with EC50 values for each compound 30-50-fold below the final concentration of PN added. Cytoprotection was correlated with a reduction in the level of measurable nitrotyrosine. In addition, we found our catalysts to be cytoprotective against endogenously generated PN in endotoxin-stimulated RAW 264.7 cells as well as in dissociated cultures of hippocampal neurons and glia that had been exposed to cytokines. Our studies thus provide compelling evidence for the involvement of peroxynitrite in cytokine-mediated cellular injury and suggest the therapeutic potential of PN decomposition catalysts in reducing cellular damage at sites of inflammation.


Subject(s)
Ferric Compounds/metabolism , Metalloporphyrins/metabolism , Nitrates/metabolism , Porphyrins/metabolism , Animals , Catalysis , Cell Death , Humans , Inflammation/metabolism , Inflammation/pathology , Nitric Oxide/metabolism , Rats , Tumor Cells, Cultured
10.
J Med Chem ; 41(9): 1361-6, 1998 Apr 23.
Article in English | MEDLINE | ID: mdl-9554868

ABSTRACT

An attractive approach to the treatment of inflammatory conditions such as osteo- and rheumatoid arthritis, inflammatory bowel disease, and sepsis is through the selective inhibition of human inducible nitric oxide synthase (hiNOS) since localized excess nitric oxide (NO) release has been implicated in the pathology of these diseases. A series of monosubstituted iminohomopiperidinium salts possessing lipophilic functionality at ring positions 3, 5, 6, and 7 has been synthesized, and series members have demonstrated the ability to inhibit the hiNOS isoform with an IC50 as low as 160 nM (7). Compounds were found that selectively inhibit hiNOS over the human endothelial constitutive enzyme (heNOS) with a heNOS/hiNOS IC50 ratio in excess of 100 and as high as 314 (9). Potencies for inhibition of hiNOS and the human neuronal constitutive enzyme (hnNOS) are comparable. Substitution in the 3 and 7 positions provides compounds that exhibit the greatest degree of selectivity for hiNOS and hnNOS over heNOS. Submicromolar potencies for 6 and 7 in a mouse RAW cell assay demonstrated the ability of these compounds to inhibit iNOS in a cellular environment. These latter compounds were also found to be orally bioavailable and efficacious due to their ability to inhibit the increase in plasma nitrite/nitrate levels in a rat LPS model.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Azepines/pharmacology , Enzyme Inhibitors/pharmacology , Imines/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Azepines/administration & dosage , Azepines/chemical synthesis , Azepines/pharmacokinetics , Biological Availability , Cell Line , Enzyme Induction/drug effects , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Humans , Imines/administration & dosage , Imines/chemical synthesis , Imines/pharmacokinetics , Inflammation/blood , Inflammation/chemically induced , Lipopolysaccharides/toxicity , Macrophages/drug effects , Macrophages/enzymology , Mice , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Rats , Rats, Inbred Lew , Recombinant Proteins/chemical synthesis , Recombinant Proteins/pharmacology , Structure-Activity Relationship
11.
Proc Natl Acad Sci U S A ; 95(5): 2659-63, 1998 Mar 03.
Article in English | MEDLINE | ID: mdl-9482943

ABSTRACT

Inflamed tissue is often characterized by the production of NO and superoxide. These radicals react at diffusion-limited rates to form the powerful oxidant peroxynitrite (PN). When protonated, PN decomposes into either nitrate or reactive intermediates capable of mediating tissue damage by oxidation of protein, lipid, and nucleic acid. We recently have identified porphyrin derivatives capable of catalyzing an increase in nitrate formation with a concomitant decrease in the HO.-like and NO2.-like reactivity of PN. Here, we present evidence for the efficacy of these PN decomposition catalysts both in vitro and in vivo. Cells in culture were protected from exogenously added PN by the catalyst 5,10,15,20-tetrakis(2,4, 6-trimethyl-3,5-disulfonatophenyl)porphyrinato iron (III), whereas free iron and the structurally related compound without iron 5,10,15, 20-tetrakis(2,4,6-trimethyl-3,5-disulfonatophenyl)porphyrin did not protect. Cytoprotection correlated well with a reduction in the nitrotyrosine content of released cytosolic proteins, a biochemical marker for PN formation. Carrageenan-induced paw edema is a model of acute inflammation in which PN may play a major role. When tested in this system, both 5,10,15,20-tetrakis(2,4,6-trimethyl-3, 5-disulfonatophenyl)porphyrinato iron (III) and 5,10,15, 20-tetrakis(N-methyl-4'-pyridyl)porphyrinato iron (III) caused a dose-dependent reduction in swelling and lactate dehydrogenase release as well as a detectable shift to nitrate formation in paw tissue. In addition, the catalysts did not elevate mean arterial pressure, suggesting a lack of interaction with NO. Taken together, our data provide compelling evidence supporting the therapeutic value of manipulating PN pharmacologically. Thus, PN decomposition catalysts may represent a unique class of anti-inflammatory agents.


Subject(s)
Edema/physiopathology , Nitrates/chemistry , Nitrates/metabolism , Porphyrins/pharmacology , Animals , Carrageenan , Catalysis , Cell Death , Cell Line , Edema/chemically induced , L-Lactate Dehydrogenase/metabolism , Macrophages , Male , Models, Chemical , Nitrates/toxicity , Nitrites/metabolism , Oxidants/chemistry , Oxidants/metabolism , Oxidants/toxicity , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
12.
Drug News Perspect ; 11(4): 204-14, 1998 May.
Article in English | MEDLINE | ID: mdl-15616662

ABSTRACT

Interest in peroxynitrite (PN) chemistry soared after recognition in 1987 of the biological role of nitric oxide, and PN has recently emerged as a possible key mediator of in vivo oxidative stress and disease. The role of PN in disease processes can be dissected both pharmacologically and biochemically, and several laboratories have examined the cellular production of peroxynitrite. In vitro demonstrations of PN-mediated cellular injury have served to strengthen the case for peroxynitrite's proposed role in the pathogenesis of human neurodegenerative disorders as examined in animal models and in diseased human tissue. Among the nervous system disorders in which PN is strongly implicated in pathology are stroke, multiple sclerosis, Alzheimer's disease, amyotrophic lateral sclerosis, and Huntington's and Parkinson's diseases. Pathologies driven by the formation of PN are amenable to pharmacological intervention at either the reactant (nitric oxide, superoxide anions) or the product (peroxynitrite). Strategies for blocking the deleterious biochemistry of peroxynitrite must aim to decrease either the flux or the intrinsic lifetime of the peroxynitrite; three particular tactics would accomplish such purposes. A novel class of antiinflammatory agents has recently been identified: PN decomposition catalysts. Identification of these catalysts offers the scientific community the opportunity to elucidate and further our understanding of the roles of peroxynitrite in animal models of diseases, which may lead to a major breakthrough in understanding the physiopathological importance of this molecule.

13.
J Neuroimmunol ; 80(1-2): 121-30, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9413267

ABSTRACT

Peroxynitrite, which is generated by the reaction of nitric oxide (NO) with superoxide, is a strong oxidant that can damage subcellular organelles, membranes and enzymes through its actions on proteins, lipids, and DNA, including the nitration of tyrosine residues of proteins. Detection of nitrotyrosine (NT) serves as a biochemical marker of peroxynitrite-induced damage. In the present studies, NT was detected by immunohistochemistry in CNS tissues from mice with acute experimental autoimmune encephalomyelitis (EAE). NT immunoreactivity was displayed by many mononuclear inflammatory cells, including CD4+ cells. It was also observed in astrocytes near EAE lesions. Immunostaining for the inducible isoform of NO synthase (iNOS) was also observed, particularly during acute EAE. These data strongly suggest that peroxynitrite formation is a major consequence of NO produced via iNOS, and implicate this powerful oxidant in the pathogenesis of EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Nitrates/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Acute Disease , Animals , Chronic Disease , Female , Immunohistochemistry , Mice , Mice, Inbred Strains , Nitric Oxide/metabolism , Nitric Oxide Synthase/analysis , Recurrence , Remission Induction , Serum Albumin, Bovine/metabolism , Tyrosine/analogs & derivatives , Tyrosine/analysis
14.
Development ; 124(18): 3587-95, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9342051

ABSTRACT

We have previously demonstrated that nitric oxide (NO)-generating compounds inhibit D. discoideum differentiation by preventing the initiation of cAMP pulses (Tao, Y., Howlett, A. and Klein, C. (1996) Cell. Signal. 8, 37-43). In the present study, we demonstrate that cells produce NO at a relatively constant rate during the initial phase of their developmental cycle. The addition of oxyhemoglobin, an NO scavenger, stimulates cell aggregation, suggesting that NO has a negative effect on the development of aggregation competence. Starvation of cells in the presence of glucose, which has been shown to prevent the initiation of cAMP pulses (Darmon, M. and Klein, C. (1978) Dev. Biol. 63, 377-389), results in an increased production of NO. The inhibition of cell aggregation by glucose treatment can be reversed by oxyhemoglobin. These findings indicate that NO is a signaling molecule for D. discoideum cells and that physiological or environmental conditions that enhance external NO levels will delay the initiation of cAMP pulses, which are essential for cell differentiation.


Subject(s)
Dictyostelium/cytology , Dictyostelium/metabolism , Nitric Oxide/physiology , Protozoan Proteins , Animals , Cell Adhesion Molecules/genetics , Cell Aggregation/drug effects , Cell Differentiation/drug effects , Culture Media , Cyclic AMP/metabolism , Dictyostelium/genetics , Dictyostelium/growth & development , Gene Expression , Glucose/pharmacology , Nitric Oxide/biosynthesis , Nitric Oxide/pharmacology , Oxyhemoglobins/pharmacology , Signal Transduction
15.
Am J Physiol ; 273(1 Pt 2): H28-37, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9249471

ABSTRACT

We previously demonstrated that inhibition of inducible nitric oxide (NO) synthase (iNOS) ameliorated acute cardiac allograft rejection. This study used a rat cardiac transplant model to characterize contractile and electrophysiological dysfunction during early acute rejection, further examine the role of NO and iNOS in this process, and determine which cells expressed iNOS during early rejection. During early acute rejection, before significant myocyte necrosis, allograft papillary muscles had reduced tension development and rates of tension development and decline during beta-adrenergic, adenylate cyclase, and calcium stimulation compared with isograft and normals [e.g., tension of 36 (allograft) vs. 73 (isograft) mN/mm2 during calcium stimulation, P < 0.001]. Allografts had resting membrane potential depolarization and reduced action potential amplitude and upstroke velocity. iNOS mRNA was expressed in infiltrating inflammatory cells but not in allograft myocytes, endothelial cells, or isografts. Corticosteroids attenuated allograft contractile and electrophysiological dysfunction and inhibited iNOS enzyme activity. Direct iNOS inhibition with aminoguanidine inhibited NO production and prevented allograft contractile and electrophysiological dysfunction (e.g., tension of 64 mN/mm2 during calcium stimulation, P < 0.001). We conclude that 1) early allograft rejection caused contractile and electrophysiological dysfunction that was largely mediated by iNOS expression in infiltrating inflammatory cells, 2) corticosteroid-mediated amelioration of allograft contractile and electrophysiological dysfunction may reflect inhibition of iNOS, and 3) iNOS inhibition may offer an alternative in management of immune-mediated myocardial dysfunction.


Subject(s)
Heart Transplantation/physiology , Myocardial Contraction , Nitric Oxide Synthase/biosynthesis , Nitric Oxide/physiology , Papillary Muscles/physiology , Action Potentials/drug effects , Adenylyl Cyclases/metabolism , Adrenal Cortex Hormones/pharmacology , Animals , Calcium/pharmacology , Cells, Cultured , Electrophysiology/methods , Guanidines/pharmacology , Heart Rate/drug effects , Heart Ventricles , In Vitro Techniques , Male , Membrane Potentials/drug effects , Methylprednisolone/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Papillary Muscles/drug effects , RNA Probes , RNA, Messenger/biosynthesis , Rats , Rats, Inbred ACI , Rats, Inbred Lew , Transcription, Genetic , Transplantation, Heterotopic , Transplantation, Homologous/physiology , Transplantation, Isogeneic/physiology
16.
Transplantation ; 63(8): 1095-101, 1997 Apr 27.
Article in English | MEDLINE | ID: mdl-9133470

ABSTRACT

BACKGROUND: We recently demonstrated that inhibition of inducible nitric oxide synthase (iNOS) ameliorated severe acute lung allograft rejection. This study used a rat lung transplant model to determine (1) the time course and cellular localization of iNOS expression during the histological progression of unmodified acute rejection and (2) whether inhibition of iNOS prevented impaired gas exchange function of the allograft lung and/or ameliorated the histological changes of acute rejection. METHODS AND RESULTS: iNOS mRNA and enzyme activity were expressed in allograft lungs during mild, moderate, and severe acute rejection, but not in normal, isograft, or allograft lungs before histological changes of mild acute rejection. iNOS expression in allografts resulted in elevated serum nitrite/nitrate levels, indicative of increased in vivo nitric oxide (NO) production. In situ hybridization demonstrated iNOS mRNA expression in infiltrating inflammatory cells, but not in allograft parenchymal cells. Allografts had significantly impaired gas exchange, which was prevented with the selective iNOS inhibitor aminoguanidine (PaO2 of 566+/-19, 76+/-22, and 504+/-105 mmHg for isograft, allograft, and aminoguanidine-treated allograft, respectively; P<0.0002). Aminoguanidine also significantly improved the histological rejection scores. CONCLUSIONS: (1) iNOS expression and increased NO production occurred during the early stages of acute rejection, persisted throughout the unmodified rejection process, and localized to infiltrating inflammatory cells, but not allograft parenchymal cells; (2) aminoguanidine ameliorated the histological and functional changes of acute rejection; and (3) increased NO production, detected by the presence of iNOS mRNA, protein, or noninvasively by measuring serum nitrite/nitrate levels, may serve as an early marker of acute allograft rejection.


Subject(s)
Lung Transplantation/immunology , Nitric Oxide Synthase/antagonists & inhibitors , Acute Disease , Animals , Gene Expression , Graft Rejection/pathology , Graft Rejection/physiopathology , In Situ Hybridization , Male , Nitric Oxide Synthase/genetics , RNA Probes , RNA, Antisense/metabolism , RNA, Messenger/metabolism , Rats , Rats, Inbred BN , Rats, Inbred F344 , Transplantation, Homologous/immunology , Transplantation, Homologous/pathology
17.
J Heart Lung Transplant ; 16(3): 334-9, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9087877

ABSTRACT

BACKGROUND: We recently demonstrated that inhibition of nitric oxide (NO) production ameliorated acute pulmonary allograft rejection. This study examined whether inducible NO synthase (iNOS) was expressed in the transplanted lung during acute rejection. METHODS: With a rat left lung transplant model, tissue from syngeneic (Fischer 344 to Fischer 344) and allogeneic (Brown Norway to Fischer 344) transplants were harvested on postoperative day 4 and analyzed for iNOS mRNA expression (ribonuclease protection assay), iNOS enzyme activity (conversion of L-[3H]-arginine to NO and L-[3H]-citrulline), and serum nitrite/nitrate levels. RESULTS: The iNOS mRNA was expressed in allograft lungs but was not detected in isografts or controls. The iNOS protein was present in allograft lungs, as demonstrated by high levels of L-[3H]-citrulline production compared with minimal iNOS enzyme activity in isograft and control lungs (10.1 +/- 2.4 vs 0.6 +/- 0.2 and 0.7 +/- 0.2 pmol L-[3H]-citrulline.mg-1.min-1, respectively; n = 6, p < 0.001). Allografts had significantly elevated systemic serum nitrite/nitrate levels compared with isografts and controls (38 +/- 6 vs 18 +/- 2 and 16 +/- 1 mumol/L, respectively; n = 6; p < 0.005). CONCLUSIONS: These results, together with our previous demonstration that iNOS inhibition ameliorated lung allograft rejection, suggest that (1) iNOS expression and increased NO production contributed to acute rejection of the transplanted lung, (2) iNOS inhibition may offer an alternative in management of acute lung allograft rejection, and (3) increased NO production, detected by the presence of iNOS mRNA or protein or noninvasively by measuring serum nitrite/nitrate levels, may serve as an early marker of acute allograft rejection.


Subject(s)
Graft Rejection/genetics , Lung Transplantation/immunology , Nitric Oxide Synthase/genetics , Animals , Enzyme Induction/genetics , Gene Expression Regulation, Enzymologic/physiology , Graft Rejection/enzymology , Lung/immunology , Male , Nitric Oxide/physiology , RNA, Messenger/genetics , Rats , Rats, Inbred BN , Rats, Inbred F344 , Transplantation, Homologous
18.
Am J Physiol ; 273(6): H2565-74, 1997 12.
Article in English | MEDLINE | ID: mdl-9435588

ABSTRACT

Tumor necrosis factor (TNF-alpha) and nitric oxide (NO) are important vasoactive mediators of septic shock. This study used a well-characterized quantitative permeation method to examine the effect of TNF-alpha and NO on systemic vascular barrier function in vivo, without confounding endotoxemia, hypotension, or organ damage. Our results showed 1) TNF-alpha reversibly increased albumin permeation in the systemic vasculature (e.g., lung, liver, brain, etc.); 2) TNF-alpha did not affect hemodynamics or blood flow or cause significant tissue injury; 3) pulmonary vascular barrier dysfunction was associated with increased lung water content and impaired oxygenation; 4) TNF-alpha caused inducible nitric oxide synthase (iNOS) mRNA expression in the lung and increased in vivo NO production; 5) selective inhibition of iNOS with aminoguanidine prevented TNF-alpha-induced lung and liver vascular barrier dysfunction; 6) aminoguanidine prevented increased tissue water content in TNF-alpha-treated lungs and improved oxygenation; and 7) nonselective inhibition of NOS with NG-monomethly-L-arginine increased vascular permeation in control lungs and caused severe lung injury in TNF-alpha-treated animals. We conclude that 1) TNF-alpha reversibly impairs vascular barrier integrity through NO-dependent and -independent mechanisms; 2) nonselective NOS inhibition increased vascular barrier dysfunction and caused severe lung injury, whereas selective inhibition of iNOS prevented impaired endothelial barrier integrity and pulmonary dysfunction; and 3) selective inhibition of iNOS may be beneficial in treating increased vascular permeability that complicates endotoxemia and cytokine immunotherapy.


Subject(s)
Capillary Permeability/drug effects , Endothelium, Vascular/physiology , Hemodynamics/physiology , Nitric Oxide Synthase/biosynthesis , Nitric Oxide/physiology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Brain/blood supply , Capillary Permeability/physiology , Endothelium, Vascular/drug effects , Guanidines/pharmacology , Hemodynamics/drug effects , Intestines/blood supply , Liver/blood supply , Lung/enzymology , Lung/pathology , Male , Muscle, Skeletal/blood supply , Nitric Oxide Synthase Type II , RNA, Messenger/biosynthesis , Rats , Rats, Inbred ACI , Recombinant Proteins/pharmacology , Regional Blood Flow/drug effects , Serum Albumin, Bovine/pharmacokinetics , Skin/blood supply , Transcription, Genetic/drug effects , omega-N-Methylarginine/pharmacology
19.
J Neuroimmunol ; 71(1-2): 145-53, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8982114

ABSTRACT

Messenger RNA encoding inducible NO synthase (iNOS) was measured by competitive reverse transcriptase polymerase chain reaction (cRT-PCR) and ribonuclease protection assays in spinal cords from mice at varying stages of experimental allergic encephalomyelitis (EAE) and from control mice. iNOS mRNA was increased in spinal cords from mice with acute EAE. cRT-PCR assays revealed a 10-20-fold increase in iNOS mRNA in spinal cords during acute EAE compared with the level observed in normal mouse spinal cords. Functional iNOS activity, as assessed by assay of calcium-independent citrulline production, was also significantly increased in spinal cords from mice with acute EAE in comparison to normal controls. The correlation of functional iNOS expression with active disease in EAE in consistent with a pathogenic role for excess NO in this model of cell-mediated central nervous system autoimmunity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/enzymology , Nitric Oxide Synthase/metabolism , Adoptive Transfer , Animals , Citrulline/metabolism , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Enzyme Induction , Female , Gene Expression , Mice , Mice, Inbred Strains , Polymerase Chain Reaction/methods , RNA, Messenger/genetics , Spinal Cord/enzymology
20.
Ann Thorac Surg ; 62(2): 378-85, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8694595

ABSTRACT

BACKGROUND: We previously demonstrated that continuous treatment with aminoguanidine, a selective inhibitor of nitric oxide production by inducible nitric oxide synthase, attenuated acute cardiac allograft rejection. METHODS: A rat transplant model was used to determine (1) when inducible nitric oxide synthase was expressed in the allograft heart during unmodified acute rejection and (2) whether pulse therapy with aminoguanidine attenuated the histologic changes of established acute rejection, in comparison with the effects of pulse therapy with corticosteroids. RESULTS: Inducible nitric oxide synthase messenger RNA and protein were expressed during early and late acute rejection. Pulse therapy with aminoguanidine inhibited nitric oxide production and attenuated the histologic changes of acute rejection, but not as effectively as corticosteroid therapy (rejection scores of 4.1 +/- 0.4, 2.5 +/- 0.9, and 1.4 +/- 0.6 on postoperative day 8, for untreated, aminoguanidine-, and dexamethasone-treated allografts, respectively (scale, 0 to 5; p < 0.05). CONCLUSIONS: (1) Inducible nitric oxide synthase expression first occurs during early acute allograft rejection and persists throughout rejection and (2) nitric oxide is an important effector molecule in acute rejection. Inducible nitric oxide synthase inhibition may offer a therapeutic adjunct in the management of acute rejection.


Subject(s)
Enzyme Inhibitors/therapeutic use , Graft Rejection/prevention & control , Guanidines/therapeutic use , Heart Transplantation/adverse effects , Nitric Oxide Synthase/antagonists & inhibitors , Transplantation, Heterologous , Animals , Dexamethasone/administration & dosage , Dexamethasone/therapeutic use , Drug Administration Schedule , Enzyme Inhibitors/administration & dosage , Gene Expression Regulation , Glucocorticoids/administration & dosage , Glucocorticoids/therapeutic use , Graft Rejection/enzymology , Graft Rejection/pathology , Guanidines/administration & dosage , Heart Transplantation/physiology , Male , Nitric Oxide Synthase/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Rats, Inbred ACI , Rats, Inbred Lew , Transplantation, Heterologous/adverse effects , Transplantation, Heterologous/physiology , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...