Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Autoimmun ; 35(4): 390-7, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20850948

ABSTRACT

Insulin is one of the earliest targeted autoantigens in the immune destruction of insulin-producing beta cells by autoreactive CD4 and CD8 T cells in type 1 diabetes. In this study, we used Non-obese diabetic (NOD) transgenic T cells engineered to express MHC class I-insulin peptide complexes linked to a T cell activation component (InsCD3-ζ), to target insulin-reactive CD8 T cells. We showed that activated, but not naïve, InsCD3-ζ CD8 T cells killed diabetogenic insulin-reactive CD8 target cells in vitro, inducing antigen-specific cell death mediated via both the release of perforin and the Fas-Fas ligand pathway. In vivo, InsCD3-ζ CD8 T cells migrated to the pancreatic lymph nodes of NOD mice after adoptive transfer. Concomitant with this, infiltration of CD8 T cells was also reduced in the pancreatic islets. Finally, in vivo, we showed that diabetes induced by adoptive transfer of insulin-reactive T cells was reduced following injection of activated InsCD3-ζ CD8 T cells. Furthermore, young NOD mice injected with InsCD3-ζ CD8 T cells developed a lower incidence and delayed onset of diabetes. Thus, using this novel system we have demonstrated that InsCD3-ζ CD8 T cells can directly kill insulin-reactive CD8 T cells in vitro and by targeting insulin-specific CD8 T cells early in the course of disease alter the progression of spontaneous diabetes in vivo in NOD mice.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/immunology , Histocompatibility Antigens Class I/metabolism , Insulin/metabolism , Peptide Fragments/metabolism , Adoptive Transfer , Animals , Autoantigens/immunology , CD3 Complex/genetics , CD3 Complex/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cytotoxicity, Immunologic/genetics , Diabetes Mellitus, Type 1/therapy , Histocompatibility Antigens Class I/genetics , Humans , Insulin/genetics , Insulin/immunology , Mice , Mice, Inbred NOD , Mice, Transgenic , Peptide Fragments/genetics , Recombinant Fusion Proteins/genetics
2.
Cell Mol Neurobiol ; 29(5): 707-17, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19330446

ABSTRACT

The potent oxidant peroxynitrite (ONOO(-)) is formed after the combination of nitric oxide with superoxide and has been closely associated with the pathology of inflammatory disease. In particular, the generation of ONOO(-) has been linked to central nervous system disorders including Alzheimer's and Parkinson's disease, multiple sclerosis and bacterial and viral meningitis. Specifically, ONOO(-) has been implicated in the loss of blood-brain barrier (BBB) integrity during neuroinflammation, but the precise mechanisms through which the molecule acts to mediate neurovascular breakdown have not been established. The disruptive effects of ONOO(-) could be mediated by either direct or indirect actions on the endothelial cells that comprise the major component of the BBB. The current study has comparatively assessed the direct toxic effects of ONOO(-) on the brain endothelial cell line, b.End3 and C6 astrocytoma and NA neuroblastoma preparations. b.End3 cells were relatively resistant to ONOO(-)-induced cell death compared with C6 and NA cultures. The indirect involvement of ONOO(-) in neuroendothelial disruption was pharmacologically determined via adhesion molecule expression and immunocompetent cell attachment to b.End3 cells. ONOO(-)-targeted drugs, including the selective free radical scavenger, uric acid, the decomposition catalyst 5,10,15,20-tetrakis (4-sulphonatophenyl) porphyrinatoiron (III) (FeTPPS) and the poly(ADP-ribose) polymerase inhibitor N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-(N,N-dimethylamino) acetamide hydrochloride (PJ34) revealed that ONOO(-) was only partly involved in E-selectin, ICAM-1 and VCAM-1 expression on b.End3 cells and also cytokine-induced T-lymphocyte attachment to the cell line. The results indicate that ONOO(-) contributes to b.End3 cell disruption but is not exclusively responsible for the breakdown of neuroendothelial function.


Subject(s)
Brain/cytology , Drug Delivery Systems , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Peroxynitrous Acid/pharmacology , Animals , Cell Adhesion/drug effects , Cell Adhesion Molecules/metabolism , Cell Death/drug effects , Cell Line , Drug Resistance/drug effects , Metalloporphyrins/pharmacology , Mice , Phenanthrenes/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Uric Acid/pharmacology
3.
Eur J Pharmacol ; 601(1-3): 88-93, 2008 Dec 28.
Article in English | MEDLINE | ID: mdl-18977216

ABSTRACT

There is increasing evidence that the oxidative radical, peroxynitrite (ONOO(-)), is involved in the pathogenesis of inflammatory diseases including multiple sclerosis and the animal counterpart, experimental autoimmune encephalomyelitis (EAE). Compounds that impede the actions of ONOO(-) have proved useful in the control of EAE. In particular, catalytic isomerisation of ONOO(-) to inactive nitrate, through the use of metalloporphyrins, curtails the cellular response to inflammatory stimuli and halts the progression of neuroinflammation during EAE. The present study examined the pharmacological effects of the metalloporphyrin and ONOO(-) decomposition catalyst 5,10,15,20-tetrakis(4-sulfonatophenyl)porphyrinatoiron(III)chloride (FeTPPS) on the acute and relapse phases of chronic relapsing (CR) EAE. Administration of FeTPPS to CR EAE-inoculated Biozzi mice commenced either therapeutically and immediately prior to the emergence of acute or relapse symptoms, or prophylactically, from the onset of remission of acute neurological signs. Drug therapy reduced acute and relapse symptoms but, and in contrast to the former phase, was of limited benefit in preventing histological changes during the latter stage of disease. In contrast, prophylactic FeTPPS was effective in limiting CNS pathology and neurological deficits. The findings confirm the inhibitory effects of FeTPPS on acute stage EAE. Moreover, the study extends previous observations by verifying compound efficacy on relapsing disease. Use of metalloporphyrins, such as FeTPPS, again highlights the important role played by ONOO(-) in the development of inflammatory diseases such as EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Metalloporphyrins/pharmacology , Peroxynitrous Acid/metabolism , Acute Disease , Animals , Catalysis , Chronic Disease , Disease Progression , Drug Administration Schedule , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Isomerism , Male , Metalloporphyrins/administration & dosage , Mice , Mice, Biozzi , Recurrence
4.
Ann N Y Acad Sci ; 1150: 54-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19120267

ABSTRACT

CD8 T cells play an important role in autoimmune diabetes development, and therefore removing these cells may protect against disease. To test this, we designed a novel method using engineered cells (InsCD3-zeta) to target insulin-specific CD8 T cells. Insulin-reactive target cells were cultured with InsCD3-zeta CD8 T cells and cytotoxicity was assessed. Activated, but not naïve, InsCD3-zeta CD8 T cells readily killed insulin-reactive target CD8 T cells. This approach to immunotarget relevant pathogenic CD8 T cells may be a therapeutic option to delay or prevent type 1 diabetes.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Cytotoxins/therapeutic use , Diabetes Mellitus, Type 1/prevention & control , Disease Models, Animal , Insulin/metabolism , Animals , CD3 Complex/genetics , CD8-Positive T-Lymphocytes/metabolism , Cell Death/drug effects , Cytotoxicity, Immunologic/genetics , Cytotoxins/pharmacology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Female , Lymphocyte Activation/drug effects , Mice , Mice, Inbred NOD , Mice, Transgenic
5.
Proc Natl Acad Sci U S A ; 104(13): 5656-61, 2007 Mar 27.
Article in English | MEDLINE | ID: mdl-17372191

ABSTRACT

Experimental allergic encephalomyelitis (EAE) is an inflammatory demyelinating disease of the CNS that is used to model certain parameters of multiple sclerosis. To establish the relative contributions of T cell reactivity, the loss of blood-brain barrier (BBB) integrity, CNS inflammation, and lesion formation toward the pathogenesis of EAE, we assessed the incidence of EAE and these parameters in mice lacking NF-kappaB, TNF-alpha, IFN-alphabeta receptors, IFN-gamma receptors, and inducible nitric oxide synthase. Although increased myelin oligodendrocyte glycoprotein-specific T cell reactivity was generally associated with a more rapid onset or increased disease severity, the loss of BBB integrity and cell accumulation in spinal cord tissues was invariably associated with the development of neurological disease signs. Histological and real-time RT-PCR analyses revealed differences in the nature of immune/inflammatory cell accumulation in the spinal cord tissues of the different mouse strains. On the other hand, disease severity during the acute phase of EAE directly correlated with the extent of BBB permeability. Thus, the loss of BBB integrity seems to be a requisite event in the development of EAE and can occur in the absence of important inflammatory mediators.


Subject(s)
Blood-Brain Barrier , Encephalomyelitis, Autoimmune, Experimental/genetics , Spinal Cord/pathology , Animals , Cell Proliferation , Central Nervous System/metabolism , Central Nervous System/pathology , Disease Models, Animal , Female , Interferon-gamma/metabolism , Male , Mice , Mice, Knockout , Permeability , Sex Factors , Spinal Cord/metabolism , T-Lymphocytes/cytology
6.
Proc Natl Acad Sci U S A ; 102(9): 3483-8, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15728348

ABSTRACT

Peroxynitrite contributes to the pathogenesis of various neurodegenerative disorders through multiple mechanisms and is thought to mediate secondary neuronal cell death after spinal cord injury (SCI). Here we establish that physiologically relevant levels of uric acid (UA), a selective inhibitor of certain peroxynitrite-mediated reactions, block the toxic effects of peroxynitrite on primary spinal cord neurons in vitro. Furthermore, administration of UA at the onset of SCI in a mouse model inhibits several pathological changes in the spinal cord including general tissue damage, nitrotyrosine formation, lipid peroxidation, activation of poly(ADP-ribose) polymerase, and neutrophil invasion. More importantly, UA treatment improves functional recovery from the injury. Taken together, our findings support the concept that peroxynitrite contributes to the pathophysiology of secondary damage after SCI. They also raise the possibility that elevating UA levels may provide a therapeutic approach for the treatment of SCI as well as other neurological diseases with a peroxynitrite-mediated pathological component.


Subject(s)
Spinal Cord Injuries/drug therapy , Uric Acid/pharmacology , Animals , Cells, Cultured , Immunohistochemistry , Lipid Peroxidation/drug effects , Male , Mice , Neurons/drug effects , Neutrophils/drug effects , Peroxidase/metabolism , Peroxynitrous Acid/antagonists & inhibitors , Spinal Cord Injuries/pathology , Uric Acid/therapeutic use
7.
J Neurotrauma ; 21(9): 1255-63, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15453994

ABSTRACT

Peroxynitrite, which has been implicated in secondary neuronal damage resulting from spinal cord injury, is capable of mediating several toxic interactions including inducing DNA strand breaks and activating the nuclear enzyme, poly (ADP-ribose) polymerase (PARP). In the present study we have tested the hypothesis that peroxynitrite-induced cell death in spinal cord injury is due to activation of PARP. Initially we examined whether peroxynitrite exerts toxic effects on primary cultures of spinal cord neurons and then determined whether the spinal cord neuronal cell death triggered by peroxynitrite was associated with PARP activation. Peroxynitrite dose-dependently reduced the viability of spinal cord neurons in vitro. Furthermore, peroxynitrite exposure markedly increased the number of DNA strand breaks in primary spinal cord neurons, resulting in activation of PARP. To identify whether PARP activation plays a direct role in peroxynitrite-induced neurotoxicity we assessed the effects of the PARP inhibitors, nicotinamide, 3-aminobenzamide and 5-iodo-6-amino-1,2 benzopyrone on cell viability in spinal cord neurons exposed to peroxynitrite. The presence of the PARP inhibitors in the cultures not only inhibited peroxynitrite-induced PARP activity in spinal cord neurons but also protected the cells from the deleterious actions of peroxynitrite. Therefore, our results demonstrate that peroxynitrite exerts toxic effects on spinal cord neurons in vitro at least in part through a PARP-dependent pathway.


Subject(s)
Neurons/enzymology , Peroxynitrous Acid/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Spinal Cord/enzymology , Animals , Cell Death/drug effects , Cell Death/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Mice , Neurons/drug effects , Spinal Cord/drug effects
8.
J Neuroimmunol ; 155(1-2): 32-42, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15342194

ABSTRACT

Urate (UA) selectively scavenges peroxynitrite-dependent radicals and suppresses CNS inflammation through effects that are manifested at the blood-brain barrier (BBB). ICAM-1 upregulation in the spinal cord tissues of myelin basic protein (MBP) immunized mice is selectively inhibited by UA treatment. In contrast, the expression of ICAM-1 and other adhesion molecules by circulating cells is unchanged. Moreover, TNF-alpha expression in the CNS tissues of MBP-immunized mice is suppressed by UA treatment but TNF-alpha-induced ICAM-1 expression on neurovascular endothelial cells is not. Therefore the effect of UA on ICAM-1 upregulation in the CNS tissues is likely due to its known contribution to the maintenance of BBB integrity in MBP-immunized mice which in turn inhibits cell invasion into the CNS and prevents TNF-alpha production in CNS tissues.


Subject(s)
Blood-Brain Barrier/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Intercellular Adhesion Molecule-1/metabolism , Spinal Cord/drug effects , Tumor Necrosis Factor-alpha/metabolism , Uric Acid/pharmacology , Animals , Blood-Brain Barrier/immunology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Female , Free Radical Scavengers/pharmacology , Free Radical Scavengers/therapeutic use , Free Radicals/immunology , Free Radicals/metabolism , Intercellular Adhesion Molecule-1/drug effects , Intercellular Adhesion Molecule-1/immunology , Mice , Myelin Basic Protein/immunology , Neurons/drug effects , Neurons/immunology , Neurons/metabolism , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase/immunology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Peroxynitrous Acid/metabolism , Spinal Cord/metabolism , Spinal Cord/physiopathology , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/immunology , Up-Regulation/drug effects , Up-Regulation/immunology , Uric Acid/therapeutic use
9.
J Pharmacol Exp Ther ; 310(3): 1053-61, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15159442

ABSTRACT

Poly(ADP-ribose) polymerase (PARP) activity has been implicated in the pathogenesis of several central nervous system (CNS) disorders. For example, the presence of extensive poly(ADP)ribosylation in CNS tissues from animals with experimental allergic encephalomyelitis (EAE) indicates that PARP activity may be involved in this inflammatory disease process. Using PJ34 [N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-N, N-dimethylacetamide.HCl], a selective PARP inhibitor, we studied the mechanisms through which PARP activity may contribute to the onset of acute EAE. PLSJL mice immunized with myelin antigens were treated with PJ34, and the effects on the progression of EAE and several other parameters relevant to the disease process were assessed. PJ34 exerted therapeutic effects at the onset of EAE that were associated with reduced CNS inflammation and the maintenance of neurovascular integrity. Expression of genes encoding the intercellular adhesion molecule-1 (ICAM-1) and the inflammatory mediators interferon-gamma, tumor necrosis factor-alpha, and inducible nitric-oxide synthase were decreased in CNS tissues from drug-treated animals. Administration of PJ34 biased the class of myelin basic protein (MBP)-specific antibodies elicited from IgG2a to IgG1 and IgG2b and modulated antigen-specific T-cell reactivity. Therefore, the mode of action of PJ34 at the onset of EAE is likely mediated by a shift in the MBP-specific immune response from a proinflammatory Th1 toward an anti-inflammatory Th2 phenotype.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Phenanthrenes/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors , Adjuvants, Immunologic/pharmacology , Animals , Blood-Brain Barrier/drug effects , Disease Models, Animal , Intercellular Adhesion Molecule-1/metabolism , Mice , Myelin Basic Protein/administration & dosage , Phenanthrenes/pharmacology , Spinal Cord Diseases/drug therapy , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Th2 Cells/drug effects , Th2 Cells/immunology , Tumor Necrosis Factor-alpha/metabolism
10.
Proc Natl Acad Sci U S A ; 100(11): 6765-70, 2003 May 27.
Article in English | MEDLINE | ID: mdl-12736376

ABSTRACT

The contribution of host factors to rabies virus (RV) transcription/replication and axonal/transsynaptic spread is largely unknown. We previously identified several host genes that are up-regulated in the mouse brain during RV infection, including neuroleukin, which is involved in neuronal growth and survival, cell motility, and differentiation, and fibroblast growth factor homologous factor 4 (FHF4), which has been implicated in limb and nervous system development. In this study, we used real-time quantitative RT-PCR to assess the expression of mRNAs specific for neuroleukin, the two isoforms of FHF4 (FHF4-1a and -1b) encoded by the FHF4 gene, and N protein of RV in neurons and astrocytes isolated by laser capture microdissection from mouse brains infected with the laboratory-adapted RV strain CVS-N2c or with a street RV of silver-haired bat origin. Differences in the gene expression patterns suggest that the capacity of RV strains to infect nonneuronal cells and differentially modulate host gene expression may be important in virus replication and spread in the CNS.


Subject(s)
Brain/metabolism , Growth Substances/genetics , Rabies virus/physiology , Animals , Base Sequence , Brain/virology , DNA Primers , Fluorescent Antibody Technique , Mice , Mice, Inbred C3H , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
Glia ; 41(2): 105-16, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12509801

ABSTRACT

Oligodendrocyte loss is a characteristic feature of several CNS disorders, including multiple sclerosis (MS) and spinal cord injury. However, the mechanisms responsible for oligodendrocyte destruction remain undefined. As recent studies have implicated peroxynitrite in the pathogenesis of both spinal cord injury and MS, we have examined whether peroxynitrite may mediate at least some of the oligodendrocyte damage and demyelination observed in these conditions. Primary rat oligodendrocytes were exposed to authentic peroxynitrite in vitro and assessed for cytotoxicity. Mitochondrial function, measured by the reduction of MTT to formazan, and mitochondrial membrane potential were used as indicators of cell viability. Cell death was quantitated by measuring either the release of lactate dehydrogenase from, or the uptake of propidium iodide into, damaged and dying cells. Peroxynitrite dose-dependently reduced the viability of primary oligodendrocytes and induced cell death. Furthermore, peroxynitrite significantly increased DNA strand breakage and the activity of poly(ADP-ribose) polymerase (PARP) in oligodendrocyte cultures. To identify whether PARP activation plays a role in peroxynitrite-induced oligodendrocyte toxicity, we examined the effects of the PARP inhibitors 3-aminobenzamide (3AB) and 5-iodo-6-amino-1,2-benzopyrone (INH(2)BP) on mitochondrial function and cell death in oligodendrocytes. The presence of 3AB and INH(2)BP did not protect oligodendrocytes from peroxynitrite-induced cytotoxicity. However, both compounds significantly reduced PARP activity in these cells. Primary oligodendrocytes generated from PARP-deficient mice were also highly susceptible to peroxynitrite-induced cell death. Therefore, our results show that peroxynitrite exerts cytotoxic effects on oligodendrocytes in vitro independently of PARP activation.


Subject(s)
Cell Death/physiology , Central Nervous System/enzymology , Multiple Sclerosis/enzymology , Oligodendroglia/enzymology , Peroxynitrous Acid/metabolism , Poly(ADP-ribose) Polymerases/deficiency , Spinal Cord Injuries/enzymology , Animals , Cell Death/drug effects , Cells, Cultured , Central Nervous System/physiopathology , Dose-Response Relationship, Drug , Female , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/enzymology , Multiple Sclerosis/physiopathology , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Oligodendroglia/drug effects , Peroxynitrous Acid/toxicity , Poly(ADP-ribose) Polymerases/drug effects , Poly(ADP-ribose) Polymerases/genetics , Spinal Cord Injuries/physiopathology
12.
Free Radic Biol Med ; 33(10): 1363-71, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12419468

ABSTRACT

Serum levels of uric acid (UA), an inhibitor of peroxynitrite- (ONOO-) related chemical reactions, became elevated approximately 30 million years ago in hominid evolution. During a similar time frame, higher mammals lost the ability to synthesize another important radical scavenger, ascorbic acid (AA), leading to the suggestion that UA may have replaced AA as an antioxidant. However, in vivo treatment with AA does not protect against the development of experimental allergic encephalomyelitis (EAE), a disease that has been associated with the activity of ONOO- and is inhibited by UA. When compared in vitro, UA and AA were found to have similar capacities to inhibit the nitrating properties of ONOO-. However UA and AA had different capacities to prevent ONOO- -mediated oxidation, especially in the presence of iron ion (Fe3+). While UA at physiological concentrations effectively blocked dihydrorhodamine-123 oxidation in the presence of Fe3+, AA did not, regardless of whether the source of ONOO- was synthetic ONOO-, SIN-1, or RAW 264.7 cells. AA also potentiated lipid peroxidation in vivo and in vitro. In conclusion, the superior protective properties of UA in EAE may be related to its ability to neutralize the oxidative properties of ONOO- in the presence of free iron ions.


Subject(s)
Ascorbic Acid/pharmacology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Molsidomine/analogs & derivatives , Tyrosine/analogs & derivatives , Uric Acid/pharmacology , Albumins/metabolism , Animals , Antioxidants/pharmacology , Ascorbic Acid/blood , Blood-Brain Barrier , Cell Line , Free Radicals , Immunohistochemistry , Iron/pharmacology , Lipid Peroxidation , Mice , Molsidomine/pharmacology , Myelin Sheath/metabolism , Nitric Oxide/metabolism , Oxygen/metabolism , Peroxynitrous Acid/antagonists & inhibitors , Rhodamines/pharmacology , Time Factors , Tyrosine/pharmacology , Uric Acid/blood
13.
Proc Natl Acad Sci U S A ; 99(25): 16303-8, 2002 Dec 10.
Article in English | MEDLINE | ID: mdl-12451183

ABSTRACT

Uric acid (UA) is a purine metabolite that selectively inhibits peroxynitrite-mediated reactions implicated in the pathogenesis of multiple sclerosis (MS) and other neurodegenerative diseases. Serum UA levels are inversely associated with the incidence of MS in humans because MS patients have low serum UA levels and individuals with hyperuricemia (gout) rarely develop the disease. Moreover, the administration of UA is therapeutic in experimental allergic encephalomyelitis (EAE), an animal model of MS. Thus, raising serum UA levels in MS patients, by oral administration of a UA precursor such as inosine, may have therapeutic value. We have assessed the effects of inosine, as well as inosinic acid, on parameters relevant to the chemical reactivity of peroxynitrite and the pathogenesis of EAE. Both had no effect on chemical reactions associated with peroxynitrite, such as tyrosine nitration, or on the activation of inflammatory cells in vitro. Moreover, when mice treated with the urate oxidase inhibitor potassium oxonate were fed inosine or inosinic acid, serum UA levels were elevated markedly for a period of hours, whereas only a minor, transient increase in serum inosine was detected. Administration of inosinic acid suppressed the appearance of clinical signs of EAE and promoted recovery from ongoing disease. The therapeutic effect on animals with active EAE was associated with increased UA, but not inosine, levels in CNS tissue. We, therefore, conclude that the mode of action of inosine and inosinic acid in EAE is via their metabolism to UA.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Inosine Monophosphate/therapeutic use , Inosine/therapeutic use , Molsidomine/analogs & derivatives , Uric Acid/metabolism , Administration, Oral , Animals , Biotransformation , Chromatography, High Pressure Liquid , Drug Evaluation, Preclinical , Encephalomyelitis, Autoimmune, Experimental/metabolism , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Humans , Inosine/pharmacokinetics , Inosine Monophosphate/pharmacokinetics , Mice , Molsidomine/pharmacology , Monocytes/drug effects , Monocytes/metabolism , Multiple Sclerosis/metabolism , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase Type II , Oxidation-Reduction , Oxidative Stress , Oxonic Acid/pharmacology , Peroxynitrous Acid/metabolism , Urate Oxidase/antagonists & inhibitors , Uric Acid/analogs & derivatives , Uric Acid/pharmacology , Xanthines/pharmacology
14.
Proc Natl Acad Sci U S A ; 99(19): 12061-6, 2002 Sep 17.
Article in English | MEDLINE | ID: mdl-12221280

ABSTRACT

Chronic inflammation results in increased nitrogen monoxide (.NO) formation and the accumulation of nitrite (NO(2-)). Neutrophils stimulated by various inflammatory mediators release myeloperoxidase to produce the cytotoxic agent hypochlorous acid (HOCl). Exposure of chondrocytic SW1353 cells to HOCl resulted in a concentration- and time-dependent loss in viability, ATP, and glutathione levels. Treatment of cells with NO(2-) but not nitrate (NO(3-)) substantially decreased HOCl-dependent cellular toxicity even when NO(2-) was added at low (microM) concentrations. In contrast, NO(2-) alone (even at 1 mM concentrations) did not affect cell viability or ATP and glutathione levels. These data suggest that NO(2-) accumulation at chronic inflammatory sites, where both HOCl and.NO are overproduced, may be cytoprotective against damage caused by HOCl. We propose that this is because HOCl is removed by reacting with NO(2-) to give nitryl chloride (NO2Cl), which is less damaging in our cell system.


Subject(s)
Hypochlorous Acid/toxicity , Nitrites/pharmacology , Adenosine Triphosphate/metabolism , Cell Line , Cell Survival/drug effects , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/metabolism , Glutathione/metabolism , Humans , Hypochlorous Acid/antagonists & inhibitors , Hypochlorous Acid/metabolism , Inflammation/etiology , Inflammation/metabolism , Inflammation/prevention & control , L-Lactate Dehydrogenase/metabolism , Nitrates/metabolism , Nitrates/pharmacology , Nitric Oxide/metabolism , Nitrites/metabolism
15.
Chronobiol Int ; 19(2): 423-39, 2002 Mar.
Article in English | MEDLINE | ID: mdl-12025934

ABSTRACT

Seven clinically healthy, nondiabetic (ND) and four Type II diabetic (D) men were assessed for circadian rhythms in oxidative "stress markers." Blood samples were collected at 3h intervals for approximately 27 h beginning at 19:00h. Urine samples were collected every 3 h beginning with the 16:00h-19:00h sample. The dark (sleep) phase of the light-dark cycle extended from 22:30h to 06:30h, with brief awakening for sampling at 01:00h and 04:00h. Subjects were offered general hospital meals at 16:30h, 07:30h, and 13:30h (2400 cal in total/24h). Serum samples were analyzed for uric acid (UA) and nitrite (NO) concentrations, and urine samples were assayed for 8-hydroxydeoxyguanosine (8-OHdG), malondialdehyde (MDA), and 8-isoprostane (ISP). Data were analyzed statistically both by the population multiple-components method and by the analysis of variance (ANOVA). The 24h mean level of UA and NO was greater in D than in ND subjects (424 vs. 338 micromol/L and 39.2 vs. 12.7 microM, respectively). A significant circadian rhythm in UA (p = 0.001) and NO (p = 0.048) was evident in ND but not in D (p = 0.214 and 0.065). A circadian rhythm (p = 0.004, amplitude = 8.6 pmol/kgbw/3h urine vol.) was also evident in urine 8-OHdG of ND but not of D. The 24h mean levels of ND and D were comparable (76.8 vs. 65.7 pmol/kgbw/3h urine vol.). No circadian rhythm by population multiple-components was evident in MDA and ISP levels of ND subjects, or in 8-OHdG, MDA, and ISP in D. However, a significant time-effect was demonstrated by ANOVA in all variables and groups. The 24h mean of MDA and ISP in D was significantly greater than in ND (214 vs. 119 nmol/3h urine vol. and 622 vs. 465 ng/3h urine vol.). The peak concentrations of the three oxidative "stress markers" in urine, like those of serum NO, occurred early in the evening in both groups of men. This observation suggests a correlation between increased oxidative damage and increased rate of anabolic-catabolic events as evidenced by similarities in the timing of peak NO production and in parameters relevant to metabolic functions.


Subject(s)
Circadian Rhythm/physiology , Deoxyguanosine/analogs & derivatives , Diabetes Mellitus, Type 2/metabolism , Dinoprost/analogs & derivatives , Oxidative Stress , 8-Hydroxy-2'-Deoxyguanosine , Adult , Aged , Biomarkers/blood , Biomarkers/urine , Case-Control Studies , Deoxyguanosine/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/urine , F2-Isoprostanes/blood , Humans , Male , Malondialdehyde/blood , Middle Aged , Nitrates/blood , Nitrites/blood , Uric Acid/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...