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










Publication year range
1.
Cancer Chemother Pharmacol ; 80(2): 421-431, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28685347

ABSTRACT

PURPOSE: Manganese porphyrins are redox-active drugs and superoxide dismutase mimics, which have been shown to chemosensitize lymphoma, a cancer which frequently occurs in dogs. This study aimed to identify critical information regarding the pharmacokinetics and toxicity of Mn(III) meso-tetrakis (N-n-butoxyetylpyridium-2-yl) porphyrin, (MnTnBuOE-2-PyP5+, MnBuOE) in dogs as a prelude to a clinical trial in canine lymphoma patients. METHODS: A single-dose pharmacokinetic (PK) study in normal dogs was performed to determine the plasma half-life (t 1/2) of MnBuOE. A dose reduction study was performed to establish the maximum tolerated dose (MTD) of MnBuOE. The safety and PK of a multi-dosing protocol was assessed. RESULTS: Peak plasma drug concentration occurred 30 min post-injection. The t 1/2 was defined as 7 h. MnBuOE induced an anaphylactic reaction and prolonged tachycardia. The MTD was defined as 0.25 mg/kg. The dogs were given MTD 3×/week for 2-3 weeks. The highest recorded tissue drug levels were in the lymph nodes (4-6 µM), followed by kidney and liver (2.5, 2.0 uM, respectively). CONCLUSIONS: We obtained critical information regarding the PK and toxicity of MnBuOE in dogs. The acute drug reaction and tachycardia post-injection have not been described in other species and may be specific to canines. The high tissue drug levels in lymph nodes have not been previously reported. MnBuOE accumulation in lymph nodes has important implications for the utility of adjuvant MnBuOE to treat lymphoma. With MnBuOE lymph node accumulation, reduction in the dose and/or administration frequency could be possible, leading to reduced toxicity.


Subject(s)
Antineoplastic Agents/administration & dosage , Kidney/metabolism , Liver/metabolism , Lymph Nodes/metabolism , Metalloporphyrins/administration & dosage , Anaphylaxis/chemically induced , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Dog Diseases/drug therapy , Dogs , Half-Life , Lymphoma/drug therapy , Lymphoma/veterinary , Male , Maximum Tolerated Dose , Metalloporphyrins/pharmacokinetics , Metalloporphyrins/toxicity , Species Specificity , Tachycardia/chemically induced , Tissue Distribution
2.
Redox Biol ; 12: 129-138, 2017 08.
Article in English | MEDLINE | ID: mdl-28231483

ABSTRACT

The signaling of reactive oxygen species (ROS) is essential for the maintenance of normal cellular function. However, whether and how ROS regulate stem cells are unclear. Here, we demonstrate that, in transgenic mice expressing the human manganese superoxide dismutase (MnSOD) gene, a scavenger of ROS in mitochondria, the number and function of mouse hematopoietic stem/progenitor cells (HSPC) under physiological conditions are enhanced. Importantly, giving MnTnBuOE-2-PyP5+(MnP), a redox- active MnSOD mimetic, to mouse primary bone marrow cells or to C57B/L6 mice significantly enhances the number of HSPCs. Mechanistically, MnP reduces superoxide to hydrogen peroxide, which activates intracellular Nrf2 signaling leading to the induction of antioxidant enzymes, including MnSOD and catalase, and mitochondrial uncoupling protein 3. The results reveal a novel role of ROS signaling in regulating stem cell function, and suggest a possible beneficial effect of MnP in treating pathological bone marrow cell loss and in increasing stem cell population for bone marrow transplantation.


Subject(s)
Hematopoietic Stem Cells/physiology , Metalloporphyrins/pharmacology , Superoxide Dismutase/metabolism , Animals , Cells, Cultured , Female , Hematopoietic Stem Cells/drug effects , Humans , Hydrogen Peroxide/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Superoxide Dismutase/genetics
3.
Free Radic Res ; 48(12): 1426-42, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25185063

ABSTRACT

Herein we have demonstrated that both superoxide dismutase (SOD) mimic, cationic Mn(III) meso-tetrakis(N-n-hexylpyridinium-2-yl)porphyrin (MnTnHex-2-PyP(5+)), and non-SOD mimic, anionic Mn(III) meso-tetrakis(4-carboxylatophenyl)porphyrin (MnTBAP(3-)), protect against oxidative stress caused by spinal cord ischemia/reperfusion via suppression of nuclear factor kappa B (NF-κB) pro-inflammatory pathways. Earlier reports showed that Mn(III) N-alkylpyridylporphyrins were able to prevent the DNA binding of NF-κB in an aqueous system, whereas MnTBAP(3-) was not. Here, for the first time, in a complex in vivo system-animal model of spinal cord injury-a similar impact of MnTBAP(3-), at a dose identical to that of MnTnHex-2-PyP(5+), was demonstrated in NF-κB downregulation. Rats were treated subcutaneously at 1.5 mg/kg starting at 30 min before ischemia/reperfusion, and then every 12 h afterward for either 48 h or 7 days. The anti-inflammatory effects of both Mn porphyrins (MnPs) were demonstrated in the spinal cord tissue at both 48 h and 7 days. The downregulation of NF-κB, a major pro-inflammatory signaling protein regulating astrocyte activation, was detected and found to correlate well with the suppression of astrogliosis (as glial fibrillary acidic protein) by both MnPs. The markers of oxidative stress, lipid peroxidation and protein carbonyl formation, were significantly reduced by MnPs. The favorable impact of both MnPs on motor neurons (Tarlov score and inclined plane test) was assessed. No major changes in glutathione peroxidase- and SOD-like activities were demonstrated, which implies that none of the MnPs acted as SOD mimic. Increasing amount of data on the reactivity of MnTBAP(3-) with reactive nitrogen species (RNS) (.NO/HNO/ONOO(-)) suggests that RNS/MnTBAP(3-)-driven modification of NF-κB protein cysteines may be involved in its therapeutic effects. This differs from the therapeutic efficacy of MnTnHex-2-PyP(5+) which presumably occurs via reactive oxygen species and relates to NF-κB thiol oxidation; the role of RNS cannot be excluded.


Subject(s)
Manganese/metabolism , Metalloporphyrins/chemistry , Metalloporphyrins/metabolism , NF-kappa B/metabolism , Reperfusion Injury/metabolism , Spinal Cord Ischemia/metabolism , Superoxide Dismutase/metabolism , Animals , Female , Manganese/chemistry , Oxidative Stress , Rats , Rats, Sprague-Dawley
4.
Free Radic Biol Med ; 72: 55-65, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24632380

ABSTRACT

Doxorubicin (DOX), one of the most effective anticancer drugs, is known to generate progressive cardiac damage, which is due, in part, to DOX-induced reactive oxygen species (ROS). The elevated ROS often induce oxidative protein modifications that result in alteration of protein functions. This study demonstrates that the level of proteins adducted by 4-hydroxy-2-nonenal (HNE), a lipid peroxidation product, is significantly increased in mouse heart mitochondria after DOX treatment. A redox proteomics method involving two-dimensional electrophoresis followed by mass spectrometry and investigation of protein databases identified several HNE-modified mitochondrial proteins, which were verified by HNE-specific immunoprecipitation in cardiac mitochondria from the DOX-treated mice. The majority of the identified proteins are related to mitochondrial energy metabolism. These include proteins in the citric acid cycle and electron transport chain. The enzymatic activities of the HNE-adducted proteins were significantly reduced in DOX-treated mice. Consistent with the decline in the function of the HNE-adducted proteins, the respiratory function of cardiac mitochondria as determined by oxygen consumption rate was also significantly reduced after DOX treatment. Treatment with Mn(III) meso-tetrakis(N-n-butoxyethylpyridinium-2-yl)porphyrin, an SOD mimic, averted the doxorubicin-induced mitochondrial dysfunctions as well as the HNE-protein adductions. Together, the results demonstrate that free radical-mediated alteration of energy metabolism is an important mechanism mediating DOX-induced cardiac injury, suggesting that metabolic intervention may represent a novel approach to preventing cardiac injury after chemotherapy.


Subject(s)
Aldehydes/metabolism , Antibiotics, Antineoplastic/toxicity , Doxorubicin/toxicity , Energy Metabolism/drug effects , Mitochondria, Heart/drug effects , Animals , Electrophoresis, Gel, Two-Dimensional , Immunoblotting , Immunoprecipitation , Lipid Peroxidation/drug effects , Male , Mice , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Oxidation-Reduction , Proteomics
5.
Oncogene ; 31(17): 2129-39, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-21909133

ABSTRACT

Manganese superoxide dismutase is a nuclear encoded primary antioxidant enzyme localized exclusively in the mitochondrial matrix. Genotoxic agents, such as ultraviolet (UV) radiation, generates oxidative stress and cause mitochondrial DNA (mtDNA) damage. The mtDNA polymerase (Polγ), a major constituent of nucleoids, is responsible for the replication and repair of the mitochondrial genome. Recent studies suggest that the mitochondria contain fidelity proteins and MnSOD constitutes an integral part of the nucleoid complex. However, it is not known whether or how MnSOD participates in the mitochondrial repair processes. Using skin tissue from C57BL/6 mice exposed to UVB radiation, we demonstrate that MnSOD has a critical role in preventing mtDNA damage by protecting the function of Polγ. Quantitative-PCR analysis shows an increase in mtDNA damage after UVB exposure. Immunofluorescence and immunoblotting studies demonstrate p53 translocation to the mitochondria and interaction with Polγ after UVB exposure. The mtDNA immunoprecipitation assay with Polγ and p53 antibodies in p53(+/+) and p53(-/-) mice demonstrates an interaction between MnSOD, p53 and Polγ. The results suggest that these proteins form a complex for the repair of UVB-associated mtDNA damage. The data also demonstrate that UVB exposure injures the mtDNA D-loop in a p53-dependent manner. Using MnSOD-deficient mice we demonstrate that UVB-induced mtDNA damage is MnSOD dependent. Exposure to UVB results in nitration and inactivation of Polγ, which is prevented by addition of the MnSOD mimetic Mn(III)TE-2-PyP(5+). These results demonstrate for the first time that MnSOD is a fidelity protein that maintains the activity of Polγ by preventing UVB-induced nitration and inactivation of Polγ. The data also demonstrate that MnSOD has a role along with p53 to prevent mtDNA damage.


Subject(s)
DNA-Directed DNA Polymerase/radiation effects , Superoxide Dismutase/physiology , Ultraviolet Rays , Animals , DNA Polymerase gamma , DNA Repair/drug effects , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/radiation effects , Metalloporphyrins/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Tumor Suppressor Protein p53/metabolism
6.
Neuroscience ; 164(2): 702-10, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19607887

ABSTRACT

Effective treatment of chronic pain with morphine is limited by decreases in the drug's analgesic action with chronic administration (antinociceptive tolerance). Because opioids are mainstays of pain management, restoring their efficacy has great clinical importance. We have recently reported that formation of peroxynitrite (ONOO(-), PN) in the dorsal horn of the spinal cord plays a critical role in the development of morphine antinociceptive tolerance and have further documented that nitration and enzymatic inactivation of mitochondrial superoxide dismutase (MnSOD) at that site provides a source for this nitroxidative species. We now report for the first time that antinociceptive tolerance in mice is also associated with the inactivation of MnSOD at supraspinal sites. Inactivation of MnSOD led to nitroxidative stress as evidenced by increased levels of products of oxidative DNA damage and activation of the nuclear factor poly (ADP-ribose) polymerase in whole brain homogenates. Co-administration of morphine with potent Mn porphyrin-based peroxynitrite scavengers, Mn(III) 5,10,15,20-tetrakis(N-ethylpyridinium-2-yl)porphyrin (MnTE-2-PyP5+) and Mn(III) 5,10,15,20-tetrakis(N-n-hexylpyridinium-2-yl)porphyrin (MnTnHex-2-PyP5+) (1) restored the enzymatic activity of MnSOD, (2) attenuated PN-derived nitroxidative stress, and (3) blocked the development of morphine-induced antinociceptive tolerance. The more lipophilic analogue, MnTnHex-2-PyP5+ was able to cross the blood-brain barrier at higher levels than its lipophylic counterpart MnTE-2-PyP5+ and was about 30-fold more efficacious. Collectively, these data suggest that PN-mediated enzymatic inactivation of supraspinal MnSOD provides a source of nitroxidative stress, which in turn contributes to central sensitization associated with the development of morphine antinociceptive tolerance. These results support our general contention that PN-targeted therapeutics may have potential as adjuncts to opiates in pain management.


Subject(s)
Analgesics/pharmacology , Brain/drug effects , Mitochondria/drug effects , Morphine/pharmacology , Peroxynitrous Acid/metabolism , Superoxide Dismutase/metabolism , Animals , Blood-Brain Barrier/drug effects , Brain/enzymology , Brain/physiology , DNA Damage/drug effects , DNA Damage/physiology , Drug Tolerance/physiology , Male , Manganese Compounds/pharmacokinetics , Manganese Compounds/pharmacology , Mice , Mice, Inbred Strains , Mitochondria/enzymology , Mitochondria/physiology , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism
7.
Neuroscience ; 153(1): 120-30, 2008 Apr 22.
Article in English | MEDLINE | ID: mdl-18353561

ABSTRACT

Alzheimer's disease (AD) is associated with beta-amyloid accumulation, oxidative stress and mitochondrial dysfunction. However, the effects of genetic mutation of AD on oxidative status and mitochondrial manganese superoxide dismutase (MnSOD) production during neuronal development are unclear. To investigate the consequences of genetic mutation of AD on oxidative damages and production of MnSOD during neuronal development, we used primary neurons from new born wild-type (WT/WT) and amyloid precursor protein (APP) (NLh/NLh) and presenilin 1 (PS1) (P264L) knock-in mice (APP/PS1) which incorporated humanized mutations in the genome. Increasing levels of oxidative damages, including protein carbonyl, 4-hydroxynonenal (4-HNE) and 3-nitrotyrosine (3-NT), were accompanied by a reduction in mitochondrial membrane potential in both developing and mature APP/PS1 neurons compared with WT/WT neurons suggesting mitochondrial dysfunction under oxidative stress. Interestingly, developing APP/PS1 neurons were significantly more resistant to beta-amyloid 1-42 treatment, whereas mature APP/PS1 neurons were more vulnerable than WT/WT neurons of the same age. Consistent with the protective function of MnSOD, developing APP/PS1 neurons have increased MnSOD protein and activity, indicating an adaptive response to oxidative stress in developing neurons. In contrast, mature APP/PS1 neurons exhibited lower MnSOD levels compared with mature WT/WT neurons indicating that mature APP/PS1 neurons lost the adaptive response. Moreover, mature APP/PS1 neurons had more co-localization of MnSOD with nitrotyrosine indicating a greater inhibition of MnSOD by nitrotyrosine. Overexpression of MnSOD or addition of MnTE-2-PyP(5+) (SOD mimetic) protected against beta-amyloid-induced neuronal death and improved mitochondrial respiratory function. Together, the results demonstrate that compensatory induction of MnSOD in response to an early increase in oxidative stress protects developing neurons against beta-amyloid toxicity. However, continuing development of neurons under oxidative damage conditions may suppress the expression of MnSOD and enhance cell death in mature neurons.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Neurons/metabolism , Oxidative Stress/genetics , Aldehydes/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Animals , Animals, Newborn , Brain/physiopathology , Cell Respiration/drug effects , Cell Respiration/physiology , Cells, Cultured , Disease Models, Animal , Humans , Membrane Potential, Mitochondrial/genetics , Metalloporphyrins/pharmacology , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondrial Diseases/physiopathology , Mutation/genetics , Neurons/drug effects , Oxidative Stress/drug effects , Presenilin-1/genetics , Protein Carbonylation/physiology , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Tyrosine/analogs & derivatives , Tyrosine/metabolism
8.
Int J Hyperthermia ; 22(4): 263-73, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16754348

ABSTRACT

PURPOSE: It has been well established that inadequate blood supply combined with high metabolic rates of oxygen consumption results in areas of low oxygen tension (<1%) within malignant tumours and that elevating tumour temperatures above 39 degrees Celsius results in significant improvement in tumour oxygenation. Macrophages play a dual role in tumour initiation and progression having both pro-tumour and anti-tumour effects. However, the response of macrophages to heat within a hypoxic environment has not yet been clearly defined. METHODS: Raw 264.7 murine macrophages were incubated under normoxia and chronic hypoxia at temperatures ranging from 37-43 degrees Celsius. Under normoxia at 41 degrees Celsius, macrophages start to release significant levels of superoxide. The combination of heat with hypoxia constitutes an additional stimulus leading to increased respiratory burst of macrophages. RESULTS: The high levels of superoxide were found to be associated with changes in macrophage production of pro-angiogenic cytokines. While hypoxia alone (37 degrees Celsius) increased levels of hypoxia inducible factor-1alpha (HIF-1alpha) in macrophages, the combination of hypoxia and mild hyperthermia (39-41 degrees Celsius) induced a strong reduction in HIF-1alpha expression. The HIF-regulated vascular endothelial growth factor (VEGF) decreased simultaneously, revealing that heat inhibits both HIF-1alpha stabilization and transcriptional activity. CONCLUSION: The data suggest that temperatures which are readily achievable in the clinic (39-41 degrees Celsius) might be optimal for maximizing hyperthermic response. At higher temperatures, these effects are reversed, thereby limiting the therapeutic benefits of more severe hyperthermic exposure.


Subject(s)
Hot Temperature , Hyperthermia, Induced , Hypoxia/metabolism , Macrophages/metabolism , Neovascularization, Physiologic/physiology , Reactive Oxygen Species/metabolism , Animals , Cell Line , Cytokines/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Mice , Neoplasms/metabolism , Neoplasms/therapy , Oxygen Consumption/physiology , Respiratory Burst/physiology , Superoxides/metabolism , Temperature , Vascular Endothelial Growth Factor A/metabolism
9.
Free Radic Res ; 39(1): 81-8, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15875815

ABSTRACT

Enhanced oxidative stress due to hyperglycemia has been implicated in diabetic complications and is considered a major cause of cell and tissue damage. The aim of the present study was to investigate whether synthetic manganese porphyrin, Mn(III) 5,10,15,20-tetrakis(N-methylpyridinium-2-yl)porphyrin (MnTM-2-PyP5+) can ameliorate diabetes-induced oxidative stress and affect life span of diabetic rats. Diabetes was induced by a single (60 mg/kg) intraperitoneal injection of streptozotocin in male Wistar rats. Oxidative stress was monitored by measuring malondialdehyde levels (MDA) in blood plasma and erythrocytes using HPLC. The antioxidant status was assessed by measuring the total radical-trapping potential (TRAP) of blood plasma. Life span of the animals was used as an indication of the overall effect of MnTM-2-PyP5+. MnTM-2-PyP5+ was administered subcutaneously at 1 mg/kg for the duration of the experiment, five times/week followed by one week of rest. Diabetes increased plasma and erythrocyte levels of MDA and decreased TRAP. MnTM-2-PyP5+ had no effect on blood glucose and glycosylated hemoglobin, but significantly increased TRAP and lowered MDA. This Mn porphyrin decreased mortality and markedly extended the life span of the diabetic animals. MnTM-2-PyP5+ suppressed diabetes-induced oxidative stress, which presumably accounts for its beneficial effect on the life span of the diabetic rats. The results indicate that Mn(III) N-alkylpyridylporphyrins can be used as potent therapeutic agents in diabetes.


Subject(s)
Diabetes Mellitus, Experimental/mortality , Manganese/chemistry , Oxidative Stress , Porphyrins/chemistry , Protoporphyrins/chemistry , Streptozocin/pharmacology , Animals , Antioxidants/pharmacology , Blood Glucose/metabolism , Chromatography, High Pressure Liquid , Erythrocytes/metabolism , Glycosylation , Hemoglobins/chemistry , Hyperglycemia , Male , Manganese/metabolism , Metalloporphyrins , Organometallic Compounds , Oxidation-Reduction , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Time Factors
10.
Proc Natl Acad Sci U S A ; 98(26): 15215-20, 2001 Dec 18.
Article in English | MEDLINE | ID: mdl-11752464

ABSTRACT

Plasma xanthine oxidase (XO) activity was defined as a source of enhanced vascular superoxide (O(2)( *-)) and hydrogen peroxide (H(2)O(2)) production in both sickle cell disease (SCD) patients and knockout-transgenic SCD mice. There was a significant increase in the plasma XO activity of SCD patients that was similarly reflected in the SCD mouse model. Western blot and enzymatic analysis of liver tissue from SCD mice revealed decreased XO content. Hematoxylin and eosin staining of liver tissue of knockout-transgenic SCD mice indicated extensive hepatocellular injury that was accompanied by increased plasma content of the liver enzyme alanine aminotransferase. Immunocytochemical and enzymatic analysis of XO in thoracic aorta and liver tissue of SCD mice showed increased vessel wall and decreased liver XO, with XO concentrated on and in vascular luminal cells. Steady-state rates of vascular O(2)( *-) production, as indicated by coelenterazine chemiluminescence, were significantly increased, and nitric oxide (( *)NO)-dependent vasorelaxation of aortic ring segments was severely impaired in SCD mice, implying oxidative inactivation of ( *)NO. Pretreatment of aortic vessels with the superoxide dismutase mimetic manganese 5,10,15,20-tetrakis(N-ethylpyridinium-2-yl)porphyrin markedly decreased O(2)( small middle dot-) levels and significantly restored acetylcholine-dependent relaxation, whereas catalase had no effect. These data reveal that episodes of intrahepatic hypoxia-reoxygenation associated with SCD can induce the release of XO into the circulation from the liver. This circulating XO can then bind avidly to vessel luminal cells and impair vascular function by creating an oxidative milieu and catalytically consuming (*)NO via O(2)( small middle dot-)-dependent mechanisms.


Subject(s)
Anemia, Sickle Cell/physiopathology , Endothelium, Vascular/physiopathology , Muscle Relaxation/physiology , Nitric Oxide/physiology , Superoxides/metabolism , Alanine Transaminase/blood , Animals , Endothelium, Vascular/metabolism , Erythrocytes/metabolism , Humans , In Vitro Techniques , Mice , Mice, Knockout , Xanthine Oxidase/blood
11.
J Neurosci ; 21(13): 4582-92, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11425886

ABSTRACT

Reactive oxygen species contribute to ischemic brain injury. This study examined whether the porphyrin catalytic antioxidant manganese (III) meso-tetrakis (N-ethylpyridinium-2-yl)porphyrin (MnTE-2-PyP(5+)) reduces oxidative stress and improves outcome from experimental cerebral ischemia. Rats that were subjected to 90 min focal ischemia and 7 d recovery were given MnTE-2-PyP(5+) (or vehicle) intracerebroventricularly 60 min before ischemia, or 5 or 90 min or 6 or 12 hr after reperfusion. Biomarkers of brain oxidative stress were measured at 4 hr after postischemic treatment (5 min or 6 hr). MnTE-2-PyP(5+), given 60 min before ischemia, improved neurologic scores and reduced total infarct size by 70%. MnTE-2-PyP(5+), given 5 or 90 min after reperfusion, reduced infarct size by 70-77% and had no effect on temperature. MnTE-2-PyP(5+) treatment 6 hr after ischemia reduced total infarct volume by 54% (vehicle, 131 +/- 60 mm(3); MnTE-2-PyP(5+), 300 ng, 60 +/- 68 mm(3)). Protection was observed in both cortex and caudoputamen, and neurologic scores were improved. No MnTE-2-PyP(5+) effect was observed if it was given 12 hr after ischemia. MnTE-2-PyP(5+) prevented mitochondrial aconitase inactivation and reduced 8-hydroxy-2'-deoxyguanosine formation when it was given 5 min or 6 hr after ischemia. In mice, MnTE-2-PyP(5+) reduced infarct size and improved neurologic scores when it was given intravenously 5 min after ischemia. There was no effect of 150 or 300 ng of MnTE-2-PyP(5+) pretreatment on selective neuronal necrosis resulting from 10 min forebrain ischemia and 5 d recovery in rats. Administration of a metalloporphyrin catalytic antioxidant had marked neuroprotective effects against focal ischemic insults when it was given up to 6 hr after ischemia. This was associated with decreased postischemic superoxide-mediated oxidative stress.


Subject(s)
Antioxidants/administration & dosage , Brain Ischemia/drug therapy , Cerebral Infarction/prevention & control , Metalloporphyrins/administration & dosage , Neuroprotective Agents/administration & dosage , Aconitate Hydratase/metabolism , Animals , Antioxidants/chemistry , Brain/blood supply , Brain/drug effects , Brain/enzymology , Brain/pathology , Brain Ischemia/etiology , Catalysis , Cerebral Infarction/etiology , DNA/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Fumarate Hydratase/metabolism , Infarction, Middle Cerebral Artery/complications , Injections, Intravenous , Injections, Intraventricular , Male , Metalloporphyrins/chemistry , Mice , Mice, Inbred C57BL , Mitochondria/enzymology , Necrosis , Neuroprotective Agents/chemistry , Oxidative Stress/drug effects , Rats , Rats, Wistar , Treatment Outcome
12.
Inorg Chem ; 40(4): 726-39, 2001 Feb 12.
Article in English | MEDLINE | ID: mdl-11225116

ABSTRACT

A manganese(III) complex of biliverdin IX dimethyl ester, (MnIIIBVDME)2, was prepared and characterized by elemental analysis, UV/vis spectroscopy, cyclic voltammetry, chronocoulometry, electrospray mass spectrometry, freezing-point depression, magnetic susceptibility, and catalytic dismuting of superoxide anion (O2.-). In a dimeric conformation each trivalent manganese is bound to four pyrrolic nitrogens of one biliverdin dimethyl ester molecule and to the enolic oxygen of another molecule. This type of coordination stabilizes the +4 metal oxidation state, whereby the +3/+4 redox cycling of the manganese in aqueous medium was found to be at E1/2 = +0.45 V vs NHE. This potential allows the Mn(III)/Mn(IV) couple to efficiently catalyze the dismutation of O2.- with the catalytic rate constant of kcat = 5.0 x 10(7) M-1 s-1 (concentration calculated per manganese) obtained by cytochrome c assay at pH 7.8 and 25 degrees C. The fifth coordination site of the manganese is occupied by an enolic oxygen, which precludes binding of NO., thus enhancing the specificity of the metal center toward O2.-. For the same reason the (MnIIIBVDME)2 is resistant to attack by H2O2. The compound also proved to be an efficient SOD mimic in vivo, facilitating the aerobic growth of SOD-deficient Escherichia coli.


Subject(s)
Biliverdine/analogs & derivatives , Biliverdine/chemistry , Manganese/chemistry , Superoxides/chemistry , Catalysis , Cytochrome c Group/metabolism , Electrochemistry , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Hydrogen Peroxide/chemistry , Magnetics , Molecular Structure , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Oxidation-Reduction , Pulse Radiolysis , Spectrometry, Mass, Electrospray Ionization , Spectrum Analysis , Superoxide Dismutase/chemistry , Superoxide Dismutase/metabolism
13.
Nitric Oxide ; 4(5): 526-33, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11020341

ABSTRACT

Reaction between NO(*) and manganese tetrakis(N-ethylpyridinium-2-yl)porphyrin (Mn(III)TE-2-PyP(5+)) was investigated at 25 degrees C. At high excess of NO(*) (1.5 mM) the reaction with the oxidized, air-stable form Mn(III)TE-2-PyP(5+) (5 microM), proceeds very slowly (t(1/2) congruent with 60 min). The presence of excess ascorbate (1 mM) produces the reduced form, Mn(II)TE-2-PyP(4+), which reacts with NO(*) stoichiometrically and in the time of mixing (k congruent with 1 x 10(6) M(-1) s(-1)). The high rate of formation and the stability of the product, Mn(II)TE-2-PyP(NO)(4+) (¿Mn(NO)¿(6)), make the reaction outcompete the reaction of NO(*) with O(2). Our in vitro measurements show a linear absorbance response upon addition of NO to a PBS, pH 7.4, solution containing an excess of ascorbate over Mn(III)TE-2-PyP(5+). Thus, the observed interactions can be the basis of a convenient and sensitive spectrophotometric assay for NO(*). Also, it may have important implications for the in vivo behavior of Mn(III)TE-2-PyP(5+) which is currently exploited as a possible therapeutic agent for various oxygen-radical related disorders.


Subject(s)
Metalloporphyrins/chemistry , Nitric Oxide/analysis , Nitroso Compounds/chemistry , Spectrophotometry/methods , Ascorbic Acid/metabolism , Calibration , Metalloporphyrins/metabolism , Molecular Structure , Nitric Oxide/chemistry , Nitroso Compounds/metabolism , Oxygen/metabolism , Sensitivity and Specificity
14.
Free Radic Biol Med ; 28(7): 1017-29, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10832063

ABSTRACT

Manganese porphyrin complexes serve to catalytically scavenge superoxide, hydrogen peroxide, and peroxynitrite. Herein, reactions of manganese 5,10,15,20-tetrakis(N-ethylpyridinium-2-yl)porphyrin (MnTE-2-PyP(5+)) with lipids and lipid hydroperoxides (LOOH) are examined. In linoleic acid and human low-density lipoprotein (LDL), MnTE-2-PyP(5+) promotes oxidative reactions when biological reductants are not present. By redox cycling between Mn(+3) and Mn(+4) forms, MnTE-2-PyP(5+) initiates lipid peroxidation via decomposition of 13(S)hydroperoxyoctadecadienoic acid [13(S)HPODE], with a second-order rate constant of 8.9 x 10(3) M(-1)s(-1)and k(cat) = 0.32 s(-1). Studies of LDL oxidation demonstrate that: (i) MnTE-2-PyP(5+) can directly oxidize LDL, (ii) MnTE-2-PyP(5+) does not inhibit Cu-induced LDL oxidation, and (iii) MnTE-2-PyP(5+) plus a reductant partially inhibit lipid peroxidation. MnTE-2-PyP(5+) (1-5 microM) also significantly inhibits FeCl(3) plus ascorbate-induced lipid peroxidation of rat brain homogenate. In summary, MnTE-2-PyP(5+) initiates membrane lipid and lipoprotein oxidation in the absence of biological reductants, while MnTE-2-PyP(5+) inhibits lipid oxidation reactions initiated by other oxidants when reductants are present. It is proposed that, as the Mn(+3) resting redox state of MnTE-2-PyP(5+) becomes oxidized to the Mn(+4) redox state, LOOH is decomposed to byproducts that propagate lipid oxidation reactions. When the manganese of MnTE-2-PyP(5+) is reduced to the +2 state by biological reductants, antioxidant reactions of the metalloporphyrin are favored.


Subject(s)
Lipids/chemistry , Lipoproteins/chemistry , Manganese/chemistry , Metalloporphyrins/chemistry , Animals , Brain Chemistry/drug effects , Catalysis , Chromatography, Thin Layer , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , Humans , Linoleic Acid/chemistry , Linoleic Acids/chemistry , Lipid Peroxidation , Lipid Peroxides/chemistry , Lipoproteins, LDL/blood , Male , Mass Spectrometry , Metalloporphyrins/pharmacology , Oxidation-Reduction , Rats , Rats, Sprague-Dawley
16.
Chem Res Toxicol ; 12(5): 442-9, 1999 May.
Article in English | MEDLINE | ID: mdl-10328755

ABSTRACT

Three isomers of manganese(III) 5,10,15, 20-tetrakis(N-methylpyridyl)porphyrin (MnTMPyP) were evaluated for their reaction with peroxynitrite. The Mn(III) complexes reacted with peroxynitrite anion with rate constants of 1.85 x 10(7), 3.82 x 10(6), and 4.33 x 10(6) M(-1) s(-1) at 37 degrees C for MnTM-2-PyP, MnTM-3-PyP, and MnTM-4-PyP, respectively, to yield the corresponding oxo-Mn(IV) complexes. Throughout the pH range from 5 to 8.5, MnTM-2-PyP reacted 5-fold faster than the other two isomers. The oxo-Mn(IV) complexes could in turn be reduced by glutathione, ascorbate, urate, or oxidize tyrosine. The rate constants for the reduction of the oxo-Mn(IV) complexes ranged from >10(7) M(-1) s(-1) for ascorbate to 10(3)-10(4) M(-1) s(-1) for tyrosine and glutathione. Cyclic voltammetry experiments show that there is no significant difference in the E1/2 of the Mn(IV)/Mn(III) couple; thus, the differential reactivity of the three isomeric complexes is interpreted in terms of electrostatic and steric effects. Micromolar concentrations of MnTM-2-PyP compete well with millimolar CO2 at reacting with ONOO-, and it can even scavenge a fraction of the ONOOCO2- that is formed. By being rapidly oxidized by ONOO- and ONOOCO2- and reduced by antioxidants such as ascorbate, urate, and glutathione, these manganese porphyrins, and especially MnTM-2-PyP, can redirect the oxidative potential of peroxynitrite toward natural antioxidants, thus protecting more critical targets such as proteins and nucleic acids.


Subject(s)
Free Radical Scavengers/chemistry , Metalloporphyrins/chemistry , Nitrates/chemistry , Oxidants/chemistry , Ascorbic Acid/chemistry , Catalysis , Glutathione/chemistry , Hydrogen-Ion Concentration , Isomerism , Kinetics , Oxidation-Reduction , Porphyrins/chemistry , Reducing Agents , Tyrosine/chemistry , Uric Acid/chemistry
17.
Free Radic Biol Med ; 26(5-6): 730-6, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10218663

ABSTRACT

The objectives of these studies were to determine whether metalloporphyrins could inhibit lipid peroxidation, characterize factors that influence their potency and compare their potency to prototypical antioxidants. Lipid peroxidation was initiated with iron and ascorbate in rat brain homogenates and the formation of thiobarbituric acid reactive species was used as an index of lipid peroxidation. Metalloporphyrins were found to be a novel and potent class of lipid peroxidation inhibitors. Inhibition of lipid peroxidation by metalloporphyrins was dependent on the transition metal ligated to the porphyrin, indicating that metal centered redox chemistry was important to the mechanism of their antioxidant activities. Manganese porphyrins with the highest superoxide dismutase (SOD) activities, MnOBTM-4-PyP and MnTM-2-PyP (charges are omitted throughout text for clarity), were the most potent inhibitors of lipid peroxidation with calculated IC50s of 1.3 and 1.0 microM, respectively. These manganese porphyrins were 2 orders of magnitude more potent than either trolox (IC50 = 204 microM) or rutin (IC50 = 112 microM). The potencies of the manganese porphyrins were related not only to their redox potentials and SOD activities, but also to other factors that may contribute to their ability to act as electron acceptors. The broad array of antioxidant activities possessed by metalloporphyrins make them attractive therapeutic agents in disease states that involve the overproduction of reactive oxygen species.


Subject(s)
Brain/metabolism , Lipid Peroxidation/drug effects , Metalloporphyrins/pharmacology , Animals , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Brain/drug effects , Chromans/pharmacology , Free Radical Scavengers/pharmacology , Kinetics , Manganese , Metalloporphyrins/chemical synthesis , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Rutin/pharmacology , Superoxide Dismutase/metabolism , Superoxide Dismutase/pharmacology , Zinc
18.
J Biol Chem ; 273(38): 24521-8, 1998 Sep 18.
Article in English | MEDLINE | ID: mdl-9733746

ABSTRACT

The ortho, meta, and para isomers of manganese(III) 5,10,15, 20-tetrakis(N-methylpyridyl)porphyrin, MnTM-2-PyP5+, MnTM-3-PyP5+, and MnTM-4-PyP5+, respectively, were analyzed in terms of their superoxide dismutase (SOD) activity in vitro and in vivo. The impact of their interaction with DNA and RNA on the SOD activity in vivo and in vitro has also been analyzed. Differences in their behavior are due to the combined steric and electrostatic factors. In vitro catalytic activities are closely related to their redox potentials. The half-wave potentials (E1/2) are +0.220 mV, +0.052 mV, and +0.060 V versus normal hydrogen electrode, whereas the rates of dismutation (kcat) are 6.0 x 10(7), 4.1 x 10(6), and 3.8 x 10(6) M-1 s-1 for the ortho, meta, and para isomers, respectively. However, the in vitro activity is not a sufficient predictor of in vivo efficacy. The ortho and meta isomers, although of significantly different in vitro SOD activities, have fairly close in vivo SOD efficacy due to their similarly weak interactions with DNA. In contrast, due to a higher degree of interaction with DNA, the para isomer inhibited growth of SOD-deficient Escherichia coli.


Subject(s)
DNA/metabolism , Metalloporphyrins/chemistry , Metalloporphyrins/metabolism , RNA/metabolism , Superoxide Dismutase/metabolism , Catalysis , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli/growth & development , Free Radical Scavengers/chemistry , Free Radical Scavengers/metabolism , Free Radical Scavengers/pharmacology , Isomerism , Kinetics , Manganese/metabolism , Metalloporphyrins/pharmacology , Molecular Structure , Oxidation-Reduction , Potentiometry , Spectrophotometry , Superoxide Dismutase/genetics
19.
Arch Biochem Biophys ; 343(2): 225-33, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9224734

ABSTRACT

Variously modified metalloporphyrins offer a promising route to stable and active mimics of superoxide dismutase (SOD). Here we explore bromination on the pyrroles as a means of increasing the redox potentials and the catalytic activities of the copper and manganese complexes of a cationic porphyrin. Mn(II) and Cu(II) octabrominated 5,10,15,20-tetrakis-(N-methylpyridinium-4-yl) porphyrin, Mn(II)OBTMPyP4+, and Cu(II)OBTMPyP4+ were prepared and characterized. The rate constants for the porphyrin-catalyzed dismutation of O2.- as determined from the inhibition of the cytochrome c reduction are k(cat) = 2.2 x 10(8) and 2.9 x 10(6) M(-1) s(-1), i.e., IC50 was calculated to be 12 nM and 0.88 microM, respectively. The metal-centered half-wave potential was E(1/2) = +0.48 V vs NHE for the manganese compound. Cu(II)OBTMPyP4+ proved to be extremely stable, while its Mn(II) analog has a moderate stability, log K = 8.08. Nevertheless, slow manganese dissociation from Mn(II)OBTMPyP4+ enabled the complex to persist and exhibit catalytic activity even at the nanomolar concentration level and at biological pH. The corresponding Mn(III)OBTMPyP5+ complex exhibited significantly increased stability, i.e., demetallation was not detected in the presence of a 400-fold molar excess of EDTA at micromolar porphyrin concentration and at pH 7.8. The beta-substituted manganese porphyrin facilitated the growth of a SOD-deficient strain of Escherichia coli when present at 0.05 microM but was toxic at 1.0 microM. The synthetic approach used in the case of manganese and copper compounds offers numerous possibilities whereby the interplay of the type and of the number of beta substituents on the porphyrin ring would hopefully lead to porphyrin compounds of increased stability, catalytic activity, and decreased toxicity.


Subject(s)
Metalloporphyrins , Superoxide Dismutase , Superoxides , Catalysis , Cytochrome c Group/metabolism , Electrochemistry , Kinetics , Metalloporphyrins/chemical synthesis , Metalloporphyrins/chemistry , Models, Molecular , Porphyrins
20.
Arch Biochem Biophys ; 324(1): 48-52, 1995 Dec 01.
Article in English | MEDLINE | ID: mdl-7503558

ABSTRACT

Detergents, such as Triton X-100, markedly increase the reduction of tetrazolium salts by xanthine oxidase plus xanthine, or by NADH. This effect of detergent, in the case of the xanthine oxidase catalyzed process, is seen aerobically but not anaerobically. Increasing the rate of accumulation of formazan, whether by increasing the concentration of the tetrazolium salt or by adding detergent, decreased susceptibility to inhibition by superoxide dismutase or by O2. These results are accommodated by a scheme of reactions the essence of which is the univalent reduction of the tetrazolium to an uncharged tetrazoinyl radical which can reduce O2 to O2- or which can partition into the detergent micelles and there dismute to generate the stable formazan.


Subject(s)
Detergents/pharmacology , Octoxynol/pharmacology , Tetrazolium Salts/metabolism , Xanthine Oxidase/metabolism , Formazans/metabolism , Models, Chemical , NAD/metabolism , Nitroblue Tetrazolium/metabolism , Oxidation-Reduction/drug effects , Oxygen/metabolism , Potentiometry , Spectrophotometry , Superoxide Dismutase
SELECTION OF CITATIONS
SEARCH DETAIL
...