Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 169(5): 1153-65, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23586466

ABSTRACT

BACKGROUND AND PURPOSE: Oestrogens can interact directly with membrane receptors and channels and can activate vascular BK(Ca) channels. We hypothesized that novel oestrogen derivatives could relax smooth muscle by an extracllular effect on the α and ß1 subunits of the BK(Ca) channel, rather than at an intracellular site. EXPERIMENTAL APPROACH: We studied the effects of novel oestrogens on the tension of pre-contracted isolated rat aortic rings, and on the electrophysiological properties of HEK 293 cells expressing the hSloα or hSloα+ß1 subunits. Two of the derivatives incorporated a quaternary ammonium side-chain making them membrane impermeable. KEY RESULTS: Oestrone, oestrone oxime and Quat DME-oestradiol relaxed pre-contracted rat aorta, but only Quat DME-oestradiol-induced relaxation was iberiotoxin sensitive. However, only potassium currents recorded in HEK 293 cells over-expressing both hSloα and hSloß1 were activated by oestrone, oestrone oxime and Quat DME-oestradiol. CONCLUSION AND IMPLICATIONS: The novel oestrogens were able to relax smooth muscle, but through different mechanisms. In particular, oestrone oxime required the presence of the endothelium to exert much of its effect, whilst Quat DME-oestradiol depended both on NO and BK(Ca) channel activation. The activation of BK(Ca) currents in HEK 293 cells expressing hSloα+ß1 by Quat DME-oestradiol is consistent with an extracellular binding site between the two subunits. The binding site resides between the extracellular N terminal of the α subunit and the extracellular loop between TM1 and 2 of the ß1 subunit. Membrane-impermeant Quat DME-oestradiol lacks an exchangeable hydrogen on the A ring obviating antioxidant activity.


Subject(s)
Aorta, Thoracic/drug effects , Estrogens/pharmacology , Large-Conductance Calcium-Activated Potassium Channels/physiology , Vasodilator Agents/pharmacology , Animals , Aorta, Thoracic/physiology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrone/pharmacology , HEK293 Cells , Humans , In Vitro Techniques , Male , Muscle Relaxation/drug effects , Muscle, Smooth, Vascular/drug effects , Oximes/pharmacology , Rats, Sprague-Dawley
2.
Int J Impot Res ; 15(6): 461-4, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14671668

ABSTRACT

The aim of the present study was to test the effects of a topical administration of a novel nitric oxide donor, linear polyethylenimine-nitric oxide/nucleophile adduct (DS1), on vaginal blood flow and hemodynamics in rats. Laser Doppler flowmetry was used to measure blood flow changes following topical application of DS1 (0.3 or 1.5 mg in 0.15 ml saline) into the vagina of anesthetized Wistar rats. In vivo hemodynamic parameters were measured with Millar-tip-catheter placed in the left ventricle. DS1 (1.5 mg) increased vaginal blood flow by 191+/-24, 226+/-22 and 166+/-23% of the baseline value (at 5, 15 and 30 min, respectively, after application) without affecting systemic blood pressure, heart rate and cardiac function. The increased vaginal blood flow following DS1 application returned to baseline between 45 and 60 min. Thus, topical application of nitric oxide donors such as DS1 may be useful for the treatment of female sexual dysfunction that develops due to an impairment of local blood flow supply to the vaginal tissue.


Subject(s)
Nitric Oxide Donors/pharmacology , Nitroso Compounds/pharmacology , Polyethyleneimine/pharmacology , Regional Blood Flow/drug effects , Vagina/blood supply , Administration, Topical , Anesthesia , Animals , Female , Rats , Rats, Wistar
3.
J Dent Res ; 82(12): 987-92, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14630900

ABSTRACT

We have investigated the role of the activation of nuclear poly(ADP-ribose) polymerase (PARP) enzyme, a mediator of downstream nitric oxide toxicity, using a combined approach of pharmacological inhibition and genetic disruption in a ligature-induced-periodontitis model in rats and mice. Immunohistochemical analysis revealed significantly increased poly(ADP-ribose) nuclear staining (indicative of PARP activation) in the subepithelial connective tissue of the ligated side compared with the non-ligated side. Ligation-induced periodontitis resulted in marked plasma extravasation in the gingivomucosal tissue and led to alveolar bone destruction compared with the non-ligated side, as measured by the Evans blue technique and by videomicroscopy, respectively. PARP inhibition with PJ34, as well as genetic PARP-1 deficiency, significantly reduced the extravasation and the alveolar bone resorption of the ligated side compared with controls. Thus, PARP activation contributes to the development of periodontal injury. Inhibition of PARP may represent a novel host response modulatory approach for the therapy of periodontitis.


Subject(s)
Periodontitis/enzymology , Poly Adenosine Diphosphate Ribose/analysis , Alveolar Bone Loss/enzymology , Alveolar Bone Loss/prevention & control , Animals , Cell Nucleus/enzymology , Connective Tissue/enzymology , Disease Models, Animal , Enzyme Activation , Gingiva/enzymology , Ligation , Male , Mice , Mice, Knockout , Mouth Mucosa/enzymology , Nuclear Proteins/analysis , Periodontitis/pathology , Periodontitis/prevention & control , Phenanthrenes/pharmacology , Plasma , Poly Adenosine Diphosphate Ribose/antagonists & inhibitors , Poly Adenosine Diphosphate Ribose/deficiency , Rats , Rats, Wistar
4.
Am J Physiol Gastrointest Liver Physiol ; 284(1): G138-44, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12388199

ABSTRACT

Inosine, a naturally occurring purine formed from the breakdown of adenosine, has recently been shown to exert powerful anti-inflammatory effects both in vivo and in vitro. This study evaluated inosine as a potential therapy for colitis. Colitis was induced in mice by the administration of dextran sulfate sodium (DSS). Oral treatment with inosine was begun either before the onset of colitis or as a posttreatment once colitis was established. Evaluation of colon damage and inflammation was determined grossly (body wt, rectal bleeding), histologically, and biochemically (colon levels of MPO, MDA, and cytokines). DSS-induced colitis significantly increased inflammatory cell infiltration into the colon. DSS-induced colitis also increased colon levels of lipid peroxidation, cytokines, and chemokines. Inosine protected the colon from DSS-induced inflammatory cell infiltration and lipid peroxidation. Inosine also partially reduced these parameters in an experimental model of established colitis. Thus inosine treatment may be a potential therapy in colitis.


Subject(s)
Colitis/drug therapy , Inosine/pharmacology , Animals , Anticoagulants , Colitis/chemically induced , Colitis/mortality , Colon/metabolism , Cytokines/metabolism , Dextran Sulfate , Disease Models, Animal , Male , Mice , Mice, Inbred BALB C , Survival Rate
5.
J Endocrinol ; 173(3): 457-64, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12065235

ABSTRACT

Insulin-dependent diabetes mellitus (IDDM) is a disease characterized by the autoimmune destruction of the pancreatic beta-cells, which requires the expression of a number of immune-related genes including major histocompatibility complex proteins, cytokines, chemokines, and cytotoxic enzymes, many of which are regulated by the transcription factor, NFkappaB. Inhibition of the entire NFkappaB family of transcription factors may be harmful, as these factors are involved in many normal physiological processes. However, identifying and targeting specific NFkappaB subunits critical for the pathogenesis of disease may prove to be valuable in designing new therapeutic strategies. To assess the potential role of the NFkappaB subunit, p50, in the development of IDDM, mice with gene disruption for NFkappaB (p50) were investigated for susceptibility to IDDM. We found that p50-deficient mice were fully resistant against multiple low-dose streptozotocin-induced diabetes, a model of diabetes with a strong autoimmune component. The site of involvement of NFkappaB (p50) lies at an early, critical juncture of immune activation and proinflammatory mediator production, because: (1) isolated islets of Langerhans from NFkappaB (p50)-deficient mice were not protected from the islet dysfunction induced by in vitro application of proinflammatory cytokines; (2) p50-deficient mice were not resistant to diabetes induced by a single high dose of streptozotocin, a model with a large oxidant component and no autoimmune involvement; and (3) diabetes induced up-regulation of nitric oxide and interleukin-12 was blocked in the p50-deficient mice. Our data suggest that NFkappaB (p50) has an essential role in the development of autoimmune diabetes. Selective therapeutic blockade of this subunit may be beneficial in preventing IDDM.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , NF-kappa B/metabolism , Animals , Cytokines/pharmacology , Disease Models, Animal , Disease Susceptibility , Drug Administration Schedule , Glucose/pharmacology , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Mice , Mice, Knockout , NF-kappa B/genetics , Protein Subunits
6.
J Pharmacol Exp Ther ; 300(3): 862-7, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11861791

ABSTRACT

Activation of the nuclear enzyme poly(ADP-ribose) polymerase (PARP) by oxidant-mediated DNA damage is an important pathway of cell dysfunction and tissue injury in conditions associated with oxidative stress. Increased oxidative stress is a major factor implicated in the cardiotoxicity of doxorubicin (DOX), a widely used antitumor anthracycline antibiotic. Thus, we hypothesized that the activation of PARP may contribute to the DOX-induced cardiotoxicity. Using a dual approach of PARP-1 suppression, by genetic deletion or pharmacological inhibition with the phenanthridinone PARP inhibitor PJ34, we now demonstrate the role of PARP in the development of cardiac dysfunction induced by DOX. PARP-1+/+ and PARP-1-/- mice received a single injection of DOX (25 mg/kg i.p). Five days after DOX administration, left ventricular performance was significantly depressed in PARP-1+/+ mice, but only to a smaller extent in PARP-1-/- ones. Similar experiments were conducted in BALB/c mice treated with PJ34 or vehicle. Treatment with a PJ34 significantly improved cardiac dysfunction and increased the survival of the animals. In addition PJ34 significantly reduced the DOX-induced increase in the serum lactate dehydrogenase and creatine kinase activities but not metalloproteinase activation in the heart. Thus, PARP activation contributes to the cardiotoxicity of DOX. PARP inhibitors may exert protective effects against the development of severe cardiac complications associated with the DOX treatment.


Subject(s)
Antibiotics, Antineoplastic , Doxorubicin , Heart Failure/chemically induced , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Acute Disease , Animals , Creatine Kinase/metabolism , Enzyme Activation/drug effects , Heart Failure/pathology , Heart Failure/physiopathology , Hemodynamics/drug effects , L-Lactate Dehydrogenase/metabolism , Male , Metalloendopeptidases/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Survival Analysis , Ventricular Function, Left/genetics
7.
J Pharmacol Exp Ther ; 300(3): 876-81, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11861793

ABSTRACT

Nicotine has been previously shown to have immunosuppressive actions. Type I diabetes is an autoimmune disease resulting from the specific destruction of the insulin-producing pancreatic beta-cells. Thus, we hypothesized that nicotine may exert protective effects against type I diabetes. The multiple low-dose streptozotocin (MLDS)-induced model and spontaneous nonobese diabetic (NOD) mouse model of type I diabetes were used to assess whether nicotine could prevent this autoimmune disease. Blood glucose levels, diabetes incidence, pancreas insulin content, and cytokine levels were measured in both models of diabetes, both to asses the level of protection exerted by nicotine and to further investigate its mechanism of action. Nicotine treatment reduced the hyperglycemia and incidence of disease in both the MLDS and NOD mouse models of diabetes. Nicotine also protected against the diabetes-induced decrease in pancreatic insulin content observed in both animal models. The pancreatic levels of the Th1 cytokines interleukin (IL)-12, IL-1, tumor necrosis factor (TNF)-alpha, and interferon (IFN)-gamma were increased in both MLDS-induced and spontaneous NOD diabetes, an effect prevented by nicotine treatment. Nicotine treatment increased the pancreatic levels of the Th2 cytokines IL-4 and IL-10. Nicotine treatment reduces the incidence of type I diabetes in two animal models by changing the profile of pancreatic cytokine expression from Th1 to Th2.


Subject(s)
Diabetes Mellitus, Experimental/prevention & control , Diabetes Mellitus, Type 1/prevention & control , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Age of Onset , Animals , Blood Glucose/metabolism , Cytokines/metabolism , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Dose-Response Relationship, Drug , Female , Insulin/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Nitrates/blood , Nitrites/blood , Pancreas/metabolism
8.
Lab Invest ; 81(12): 1683-92, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11742038

ABSTRACT

The proinflammatory cytokines, interleukin-1beta (IL-1beta), tumor necrosis factor alpha (TNFalpha), and interferon gamma (IFNgamma), are cytotoxic to pancreatic islet beta cells, possibly by inducing nitric oxide and/or oxygen radical production in the beta cells. Peroxynitrite, the reaction product of nitric oxide and the superoxide radical, is a strong oxidant and cytotoxic mediator; therefore, we hypothesized that peroxynitrite might be a mediator of cytokine-induced islet beta-cell destruction. To test this hypothesis we incubated islets isolated from human pancreata with the cytokine combination of IL-1beta, TNFalpha, and IFNgamma. We found that these cytokines induced significant increases in nitrotyrosine, a marker of peroxynitrite, in islet beta cells, and the increase in nitrotyrosine preceded islet-cell destruction. Peroxynitrite mimicked the effects of cytokines on nitrotyrosine formation and islet beta-cell destruction. L-N(G)-monomethyl arginine, an inhibitor of nitric oxide synthase, prevented cytokine-induced nitric oxide production but not hydrogen peroxide production, nitrotyrosine formation, or islet beta-cell destruction. In contrast, guanidinoethyldisulphide, an inhibitor of inducible nitric oxide synthase and scavenger of peroxynitrite, prevented cytokine-induced nitric oxide and hydrogen peroxide production, nitrotyrosine formation, and islet beta-cell destruction. These results suggest that cytokine-induced peroxynitrite formation is dependent upon increased generation of superoxide (measured as hydrogen peroxide) and that peroxynitrite is a mediator of cytokine-induced destruction of human pancreatic islet beta cells.


Subject(s)
Cytokines/pharmacology , Islets of Langerhans/drug effects , Peroxynitrous Acid/physiology , Tyrosine/analogs & derivatives , Cell Death , Drug Combinations , Guanidines/pharmacology , Humans , Immunohistochemistry/methods , In Vitro Techniques , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Islets of Langerhans/physiopathology , Tumor Necrosis Factor-alpha/pharmacology , Tyrosine/metabolism
9.
Am J Respir Crit Care Med ; 164(7): 1213-20, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11673212

ABSTRACT

Inosine is a naturally occurring purine formed from the breakdown of adenosine. Here we have evaluated the effects of inosine in a murine model of polymicrobial sepsis induced by cecal ligation and puncture (CLP). Mice subjected to CLP were treated with either inosine (100 mg/kg, intraperitoneally) or vehicle 1 h before and 6 h after CLP. After 12 h tumor necrosis factor alpha, interleukin 6 (IL-6), and IL-10 were measured in plasma. Biochemical markers of organ damage, liver NAD+/NADH (indicator of the mitochondrial redox state), plasma nitrate, tissue myeloperoxidase (MPO, indicator of neutrophil accumulation) and malondialdehyde (MDA, indicator of lipid peroxidation), liver and lung chemokines (macrophage inflammatory protein 1alpha [MIP-1alpha] and MIP-2), and ex vivo vascular reactivity in aortic rings were also measured. Mice treated with inosine had significantly lower levels of circulating cytokines. Organ damage was significantly reduced by inosine treatment, which was associated at the tissue level with an increased hepatic NAD+/NADH ratio, decreased MPO activity in the lung, reduced MDA formation in the gut and liver, and decreased MIP-1alpha and MIP-2 in the lung and liver. Furthermore, inosine significantly improved endothelium-dependent relaxant responses of aortic rings. These effects were associated with significant improvement of the survival of CLP mice treated with inosine, an effect that was still observed when inosine treatment was delayed 1 h after CLP, especially when it was associated with appropriate antibiotic treatment. Thus, inosine reduced systemic inflammation, organ damage, tissue dysoxia, and vascular dysfunction, resulting in improved survival in septic shock.


Subject(s)
Inflammation/drug therapy , Inosine/therapeutic use , Shock, Septic/drug therapy , Shock, Septic/mortality , Animals , Biomarkers/blood , Cecum , Cytokines/blood , Inflammation/blood , Inflammation/etiology , Ligation , Male , Mice , Mice, Inbred BALB C , Punctures , Shock, Septic/blood , Shock, Septic/etiology , Survival Rate
10.
J Autoimmun ; 16(4): 449-55, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11437493

ABSTRACT

Peroxynitrite (ONOO(-)) is a highly reactive oxidant produced by the interaction of the free radicals superoxide (O*-2) and nitric oxide (NO(*)). In a previous study, we found that peroxynitrite is formed in islet beta-cells of nonobese diabetic (NOD) mice. Here, we report that guanidinoethyldisulphide (GED), a selective inhibitor of inducible nitric oxide synthase (iNOS) and scavenger of peroxynitrite prevents diabetes in NOD mice. GED treatment of female NOD mice, starting at age 5 weeks, delayed diabetes onset (from age 12 to 22 weeks) and significantly decreased diabetes incidence at 30 weeks (from 80% to 17%). GED did not prevent pancreatic islet infiltration by leukocytes; however, beta-cells that stained positive for nitrotyrosine (a marker of peroxynitrite) were significantly decreased in islets of GED-treated mice (1+/-1%) compared with vehicle-treated mice (30+/-9%). In addition, GED significantly inhibited nitric oxide and nitrotyrosine formation and decreased destruction of beta-cells in NOD mouse islets incubated in vitro with the combination of proinflammatory cytokines interleukin 1-beta (IL-1beta), tumour necrosis factor-alpha (TNF-alpha) and interferon-gamma (IFN-gamma). These findings indicate that both superoxide and nitric oxide radicals contribute to islet beta-cell destruction in autoimmune diabetes via peroxynitrite formation in the beta-cells.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Guanidines/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Oxidants/metabolism , Peroxynitrous Acid/metabolism , Animals , Cells, Cultured , Diabetes Mellitus, Type 1/metabolism , Enzyme Inhibitors/administration & dosage , Female , Free Radical Scavengers/administration & dosage , Guanidines/administration & dosage , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Male , Mice , Mice, Inbred NOD , Nitric Oxide Synthase Type II , Pancreas/metabolism , Pancreas/pathology
11.
Br J Pharmacol ; 133(6): 909-19, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11454665

ABSTRACT

Activation of poly(ADP-ribose) synthetase (PARS, also termed polyADP-ribose polymerase or PARP) has been proposed as a major mechanism contributing to beta-cell destruction in type I diabetes. In the present study, we have investigated the role of PARS in mediating the induction of diabetes and beta-cell death in the multiple-low-dose-streptozotocin (MLDS) model of type I diabetes. Mice genetically deficient in PARS were found to be less sensitive to MLDS than wild type mice, with a lower incidence of diabetes and reduced hyperglycemia. A potent inhibitor of PARS, 5-iodo-6-amino-1,2-benzopyrone (INH(2)BP), was also found to protect mice from MLDS and prevent beta-cell loss, in a dose-dependent manner. Paradoxically, in the PARS deficient mice, the compound increased the onset of diabetes. In vitro the cytokine combination; interleukin-1beta, tumor necrosis factor-alpha and interferon-gamma inhibited glucose-stimulated insulin secretion from isolated rat islets of Langerhans and decreased RIN-5F cell viability. The PARS inhibitor, INH(2)BP, protected both the rat islets and the beta-cell line, RIN-5F, from these cytokine-mediated effects. These protective effects were not mediated by inhibition of cytokine-induced nitric oxide formation. Inhibition of PARS by INH(2)BP was unable to protect rat islet cells from cytokine-mediated apoptosis. Cytokines, peroxynitrite and streptozotocin were all shown to induce PARS activation in RIN-5F cells, an effect suppressed by INH(2)BP. The present study provides evidence for in vivo PARS activation contributing to beta-cell damage and death in the MLDS model of diabetes, and indicates a role for PARS activation in cytokine-mediated depression of insulin secretion and cell viability in vitro.


Subject(s)
Coumarins/pharmacology , Diabetes Mellitus, Experimental/prevention & control , Enzyme Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Apoptosis/drug effects , Blood Glucose/drug effects , Blood Glucose/metabolism , Cell Survival/drug effects , Cytokines/pharmacology , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Dose-Response Relationship, Drug , Female , Genotype , In Vitro Techniques , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mutation , Nitrates/pharmacology , Nitric Oxide/metabolism , Poly(ADP-ribose) Polymerases/genetics , Rats , Severity of Illness Index , Streptozocin/administration & dosage , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/enzymology
12.
Biochim Biophys Acta ; 1539(3): 233-42, 2001 Jun 20.
Article in English | MEDLINE | ID: mdl-11420121

ABSTRACT

The amiloride-inhibitable Na(+)/H(+) antiporter plays an important role in macrophage activation. The intracellular pathways leading to interleukin (IL)-12 p40 production by activated macrophages are incompletely understood. In the present study, we examined the contribution of the Na(+)/H(+) antiporter to the production of IL-12 p40. Amiloride or its analogs decreased the production of IL-12 p40 in macrophages stimulated with bacterial lipopolysaccharide and interferon-gamma. The order of potency of amiloride analogs was consistent with the proposition that the effect of amiloride is mediated by the inhibition of the Na(+)/H(+) antiporter. The effect of amiloride was post-transcriptional, as IL-12 p40 mRNA levels induced by lipopolysaccharide and interferon-gamma were not affected by this inhibitor. Furthermore, the inhibitory effect of amiloride on IL-12 p40 production was not a result of interference with the activation of the p38 and p42/44 mitogen-activated protein kinases or c-Jun kinase. In summary, the production of IL-12 p40 requires a functional Na(+)/H(+) antiporter.


Subject(s)
Amiloride/pharmacology , Interleukin-12/metabolism , Macrophages, Peritoneal/drug effects , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Amiloride/analogs & derivatives , Animals , Chemokine CCL4 , Chemokine CXCL2 , Chemokines/metabolism , Diuretics/pharmacology , Drug Interactions , Enzyme Activation/drug effects , Gene Silencing , JNK Mitogen-Activated Protein Kinases , Lipopolysaccharides/pharmacology , Macrophage Inflammatory Proteins/metabolism , Macrophages, Peritoneal/metabolism , Male , Methacrylates/pharmacology , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Sodium-Hydrogen Exchangers/metabolism , p38 Mitogen-Activated Protein Kinases
13.
Nat Med ; 7(1): 108-13, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11135624

ABSTRACT

Diabetic patients frequently suffer from retinopathy, nephropathy, neuropathy and accelerated atherosclerosis. The loss of endothelial function precedes these vascular alterations. Here we report that activation of poly(ADP-ribose) polymerase (PARP) is an important factor in the pathogenesis of endothelial dysfunction in diabetes. Destruction of islet cells with streptozotocin in mice induced hyperglycemia, intravascular oxidant production, DNA strand breakage, PARP activation and a selective loss of endothelium-dependent vasodilation. Treatment with a novel potent PARP inhibitor, starting after the time of islet destruction, maintained normal vascular responsiveness, despite the persistence of severe hyperglycemia. Endothelial cells incubated in high glucose exhibited production of reactive nitrogen and oxygen species, consequent single-strand DNA breakage, PARP activation and associated metabolic and functional impairment. Basal and high-glucose-induced nuclear factor-kappaB activation were suppressed in the PARP-deficient cells. Our results indicate that PARP may be a novel drug target for the therapy of diabetic endothelial dysfunction.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Endothelium, Vascular/physiopathology , Poly(ADP-ribose) Polymerases/metabolism , Animals , Diabetes Mellitus, Experimental/enzymology , Endothelium, Vascular/enzymology , Enzyme Activation , Glucose/pharmacology , Immunohistochemistry , In Vitro Techniques , Mice , NF-kappa B/metabolism
14.
Inflamm Res ; 50(11): 561-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11766996

ABSTRACT

OBJECTIVE AND DESIGN: Oxygen- and nitrogen-derived free radicals and oxidants play an important role in the pathogenesis of various forms of inflammation. Recent work emphasizes the importance of oxidant-induced DNA strand breakage and activation of the nuclear enzyme poly(ADP-ribose) polymerase (PARP) in the pathogenesis of various inflammatory diseases. We have recently demonstrated the efficacy of PJ34, a novel, potent phenanthridinone derivative PARP inhibitor, in rodent models of diabetic vascular dysfunction and stroke. Here we tested the efficacy of PARP inhibition in various models of local inflammation in rodents. MATERIALS AND METHODS: PJ34 (at doses of 0.03-30 mg/kg) was tested in rats and mice subjected to standard models of inflammation, with relevant parameters of inflammation measured using standard methods. RESULTS: PJ34 treatment (s.c, i.p. and i.v.) dose-dependently suppressed neutrophil infiltration and nitric oxide (but not KC and IL-1beta) production in peritonitis. In a model of systemic endotoxemia, PJ34 pretreatment significantly reduced plasma levels of TNF-alpha, IL-1beta and nitrite/nitrate (breakdown products of nitric oxide) production. PJ34 treatment (oral gavage) induced a significant suppression of the inflammatory response in dextran sulfate colitis, multiple low dose streptozotocin diabetes and cyclophosphamide-accelerated autoimmune diabetes in the non-obese diabetic mice, and reduced the degree of mononuclear cell infiltration into the iris in an endotoxin-induced uveitis model. Delaying the start of PJ34 administration in the colitis model conferred significant protective effects, while in the arthritis model the post-treatment paradigm lacked protective effects. CONCLUSIONS: PJ34 provides significant, dose-dependent, anti-inflammatory effects in a variety of local inflammation models. Some of its actions are maintained in the post-treatment regimen and/or after discontinuation of treatment. We conclude that PARP inhibition offers a powerful means for reducing the severity of various forms of local inflammatory responses.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Enzyme Inhibitors/pharmacology , Phenanthrenes/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Adenosine Triphosphate/metabolism , Animals , Arthritis/drug therapy , Colitis/drug therapy , Collagen/immunology , Diabetes Mellitus, Experimental/drug therapy , Dose-Response Relationship, Drug , Endotoxemia/drug therapy , Male , Mice , Mice, Inbred DBA , NAD/metabolism , Peritonitis/drug therapy , Uveitis/drug therapy
15.
FASEB J ; 14(13): 2065-74, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11023991

ABSTRACT

Interleukin 12 (IL-12) is a crucial cytokine in the regulation of T helper 1 vs. T helper 2 immune responses. In the present study, we investigated the effect of the endogenous purine nucleoside adenosine on the production of IL-12. In mouse macrophages, adenosine suppressed IL-12 production. Although the order of potency of adenosine receptor agonists suggested the involvement of A2a receptors, data obtained with A2a receptor-deficient mice showed that the adenosine suppression of IL-12 and even TNF-alpha production is only partly mediated by A2a receptor ligation. Studies with adenosine receptor antagonists or the adenosine uptake blocker dipyridamole showed that adenosine released endogenously also decreases IL-12. Although adenosine increases IL-10 production, the inhibition of IL-12 production is independent of the increased IL-10. The mechanism of action of adenosine was not associated with alterations of the activation of the p38 and p42/p44 mitogen-activated protein kinases or the phosphorylation of the c-Jun terminal kinase. Adenosine failed to affect steady-state levels of either IL-12 p35 or p40 mRNA, but augmented IL-10 mRNA levels. In summary, adenosine inhibits IL-12 production via various adenosine receptors. These results support the notion that adenosine-based therapies might be useful in certain autoimmune and/or inflammatory diseases.


Subject(s)
Adenosine/pharmacology , Interleukin-12/biosynthesis , Macrophages/drug effects , Receptors, Purinergic P1/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Interleukin-10/biosynthesis , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Mitogen-Activated Protein Kinases/metabolism , Purinergic P1 Receptor Agonists , Purinergic P1 Receptor Antagonists , Receptor, Adenosine A2A , Receptors, Purinergic P1/deficiency , Receptors, Purinergic P1/genetics
16.
Proc Natl Acad Sci U S A ; 97(18): 10203-8, 2000 Aug 29.
Article in English | MEDLINE | ID: mdl-10954738

ABSTRACT

Hemorrhagic shock (HS) and resuscitation leads to widespread production of oxidant species. Activation of the enzyme poly(ADP-ribose) polymerase (PARP) has been shown to contribute to cell necrosis and organ failure in various disease conditions associated with oxidative stress. We tested the hypothesis whether PARP activation plays a role in the multiple organ dysfunction complicating HS and resuscitation in a murine model of HS and resuscitation by using mice genetically deficient in PARP (PARP(-/-)) and their wild-type littermates (PARP(+/+)). Animals were bled to a mean blood pressure of 45 mmHg (1 mmHg = 133 Pa) and resuscitated after 45 min with isotonic saline (2x volume of shed blood). There was a massive activation of PARP, detected by poly(ADP-ribose) immunohistochemistry, which localized to the areas of the most severe intestinal injury, i.e., the necrotic epithelial cells at the tip of the intestinal villi, and colocalized with tyrosine nitration, an index of peroxynitrite generation. Intestinal PARP activation resulted in gut hyperpermeability, which developed in PARP(+/+) but not PARP(-/-) mice. PARP(-/-) mice were also protected from the rapid decrease in blood pressure after resuscitation and showed an increased survival time, as well as reduced lung neutrophil sequestration. The beneficial effects of PARP suppression were not related to a modulation of the NO pathway nor to a modulation of signaling through IL-6, which similarly increased in both PARP(+/+) and PARP(-/-) mice exposed to HS. We propose that PARP activation and associated cell injury (necrosis) plays a crucial role in the intestinal injury, cardiovascular failure, and multiple organ damage associated with resuscitated HS.


Subject(s)
Hemodynamics , Intestinal Mucosa/pathology , Muscle, Smooth, Vascular/physiopathology , Poly(ADP-ribose) Polymerases/metabolism , Shock, Hemorrhagic/genetics , Acetylcholine/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Aorta, Thoracic/physiopathology , Blood Pressure , Blood Volume , Dinoprost/pharmacology , Enzyme Activation , In Vitro Techniques , Intestinal Mucosa/enzymology , Liver/enzymology , Liver/pathology , Male , Mice , Mice, Knockout , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Peroxidase/metabolism , Poly(ADP-ribose) Polymerases/deficiency , Poly(ADP-ribose) Polymerases/genetics , Resuscitation , Shock, Hemorrhagic/physiopathology , Shock, Hemorrhagic/therapy , Sodium Chloride/therapeutic use , Tyrosine/analogs & derivatives , Tyrosine/metabolism
17.
J Immunol ; 164(2): 1013-9, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10623851

ABSTRACT

Extracellular purines, including adenosine and ATP, are potent endogenous immunomodulatory molecules. Inosine, a degradation product of these purines, can reach high concentrations in the extracellular space under conditions associated with cellular metabolic stress such as inflammation or ischemia. In the present study, we investigated whether extracellular inosine can affect inflammatory/immune processes. In immunostimulated macrophages and spleen cells, inosine potently inhibited the production of the proinflammatory cytokines TNF-alpha, IL-1, IL-12, macrophage-inflammatory protein-1alpha, and IFN-gamma, but failed to alter the production of the anti-inflammatory cytokine IL-10. The effect of inosine did not require cellular uptake by nucleoside transporters and was partially reversed by blockade of adenosine A1 and A2 receptors. Inosine inhibited cytokine production by a posttranscriptional mechanism. The activity of inosine was independent of activation of the p38 and p42/p44 mitogen-activated protein kinases, the phosphorylation of the c-Jun terminal kinase, the degradation of inhibitory factor kappaB, and elevation of intracellular cAMP. Inosine suppressed proinflammatory cytokine production and mortality in a mouse endotoxemic model. Taken together, inosine has multiple anti-inflammatory effects. These findings, coupled with the fact that inosine has very low toxicity, suggest that this agent may be useful in the treatment of inflammatory/ischemic diseases.


Subject(s)
Cytokines/antagonists & inhibitors , Immunosuppressive Agents/pharmacology , Inflammation Mediators/antagonists & inhibitors , Inosine/pharmacology , Lipopolysaccharides/toxicity , Protein Processing, Post-Translational/drug effects , Shock, Septic/immunology , Shock, Septic/prevention & control , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Chemokines/antagonists & inhibitors , Chemokines/biosynthesis , Cytokines/biosynthesis , Enzyme Activation/drug effects , Enzyme Activation/immunology , I-kappa B Proteins/metabolism , Inflammation Mediators/metabolism , Injections, Intraperitoneal , Inosine/administration & dosage , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , JNK Mitogen-Activated Protein Kinases , Macrophage Activation/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Protein Processing, Post-Translational/immunology , Purinergic P1 Receptor Agonists , Receptors, Purinergic P1/physiology , Shock, Septic/etiology , Shock, Septic/pathology , Th1 Cells/drug effects , Th1 Cells/metabolism
18.
Cytokine ; 11(8): 585-92, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10433805

ABSTRACT

Nitric oxide synthase, induced by cytokines in insulin-containing cells, produces nitric oxide which inhibits function and may promote cell killing. Since glucagon was shown to prevent inducible nitric oxide synthase (iNOS) expression in rat hepatocytes it was of interest to examine the action of glucagon (and cyclic AMP) on iNOS induction in insulin-producing cells. Cultured RIN5F cells and primary rat and human islets of Langerhans were treated with interleukin 1beta (IL-1beta) or a combination of cytokines, and were co-treated or pre-treated with glucagon. In RIN5F cells, the activity of iNOS induced by IL-1beta (10 pM, 24 h), was significantly reduced by glucagon (1000 nM), which raises cyclic AMP, and by forskolin (1-10 microM), a non specific activator of adenylate cyclase. Glucagon and forskolin also decreased iNOS expression in RIN5F cells, and rat and human islets, as shown by Western blotting. The inhibitory action of IL-1beta (100 pM, 24 h) on rat islet insulin secretion was partially reversed by 1-h pre-treatment with glucagon (10-1000 nM), while the contrasting stimulatory effect of 48-h treatment with cytokines on insulin secretion from human islets was similarly prevented by glucagon (1000 nM) pre-treatment. These results suggest that glucagon inhibits iNOS expression in insulin-containing cells and imply that glucagon could modulate the inhibitory effects of cytokines.


Subject(s)
Cytokines/pharmacology , Gene Expression Regulation, Enzymologic/physiology , Glucagon/pharmacology , Insulin/metabolism , Interleukin-1/pharmacology , Islets of Langerhans/physiology , Nitric Oxide Synthase/biosynthesis , Animals , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/metabolism , Enzyme Induction , Female , Gene Expression Regulation, Enzymologic/drug effects , Humans , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Kinetics , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Rats , Rats, Sprague-Dawley , Time Factors , Tumor Cells, Cultured
19.
FEBS Lett ; 417(2): 235-8, 1997 Nov 10.
Article in English | MEDLINE | ID: mdl-9395303

ABSTRACT

We have previously observed that treatment of rat islets of Langerhans with interleukin-1beta for 12 h results in nitric oxide-dependent inhibition of insulin secretion, while 48 h treatment increased rates of islet cell death by apoptosis. Here, we demonstrate that interleukin-1beta-mediated nitric oxide formation and inhibition of insulin secretion are significantly reduced by 24 h pretreatment of rat islets of Langerhans with insulin-like growth factor I (IGF-I). IGF-I decreased cytokine induction of nitric oxide synthase in islets. Use of an arginine analogue in culture or IGF-I pretreatment of islets were also effective in protecting islets against cytokine-mediated apoptotic cell death. We conclude that IGF-I antagonises inhibitory and cytotoxic effects of cytokines in rat islets.


Subject(s)
Apoptosis/drug effects , Insulin-Like Growth Factor I/pharmacology , Insulin/metabolism , Interleukin-1/pharmacology , Islets of Langerhans/metabolism , Nitric Oxide Synthase/biosynthesis , Animals , Animals, Newborn , Enzyme Induction/drug effects , Female , Insulin Secretion , Islets of Langerhans/drug effects , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley , Secretory Rate/drug effects , Tumor Necrosis Factor-alpha/pharmacology
20.
Cytokine ; 9(8): 570-6, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9245484

ABSTRACT

Induction of nitric oxide synthase and generation of nitric oxide in pancreatic islet beta-cells may mediate cytokine-induced dysfunction leading to insulin-dependent diabetes mellitus. Nitric oxide generation can be regulated by availability of arginine substrate which, in turn, may be affected by substrate utilization in competing pathways such as the arginase-catalysed formation of ornithine and urea. In this study we have investigated the activity of arginase in the rat insulinoma-derived cell line RINm5F and the effect on this of interleukin 1beta, the nitric oxide synthase reaction intermediate NG-hydroxy-l-arginine and the nitric oxide-generating compounds 3-morpholinosydnonimine and S-nitrosoglutathione. Cytosols from RINm5F cells treated with or without interleukin 1beta (0.1nM, 18h) were incubated (45min, 37 degrees C) with [U-14C]arginine. Radiolabelled products ([14C]citrulline from nitric oxide synthase, [14C]ornithine and [14C]urea from arginase) were separated by high-performance liquid chromatography or ion-exchange chromatography. Interleukin 1beta increased citrulline production (from 0.01+/-0.002 to 0.58+/-0.03 pmol/microg cell protein), indicating induction of nitric oxide synthase, and significantly decreased production of both ornithine (from 4.60+/-0.20 to 3.40+/-0.20 pmol/microg) and urea (0.93+/-0.05 to 0.69+/-0.04 pmol/microg) (P<0.001), indicating decreased activity of arginase. Arginase was significantly inhibited by NG-hydroxy-l-arginine (IC50=50 microM), S-nitrosoglutathione (500 microM: 69+/-7% of control) and 3-morpholinosydnonimine (1 mM: 57+/-7% of control) (P<0.05). We conclude that during cytokine-directed beta-cell assault nitric oxide synthase-catalysed production of NG-hydroxy-l-arginine and nitric oxide may inhibit arginase thereby increasing the availability of arginine for nitric oxide production.


Subject(s)
Arginase/antagonists & inhibitors , Interleukin-1/pharmacology , Animals , Arginine/isolation & purification , Citrulline/isolation & purification , Enzyme Inhibitors/pharmacology , Glutathione/analogs & derivatives , Glutathione/pharmacology , Insulinoma , Islets of Langerhans/drug effects , Islets of Langerhans/enzymology , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , Nitric Oxide Synthase/metabolism , Nitroso Compounds/pharmacology , Ornithine/isolation & purification , Rats , S-Nitrosoglutathione , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...