Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Free Radic Res ; 47(6-7): 507-10, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23566300

ABSTRACT

Lipid peroxidation generates reactive aldehydes, most notably hydroxynonenal (HNE), which covalently binds amino acid residue side chains leading to protein inactivation and insolubility. Specific adducts of lipid peroxidation have been demonstrated to be intimately associated with pathological lesions of Alzheimer's disease (AD), suggesting that oxidative stress is a major component in the disease. Here, we examined the HNE-cross-linking modifications by using an antibody specific for a lysine-lysine cross-link. Since in a prior study we noted no immunolabeling of neuritic plaques or neurofibrillary tangles but instead found strong labeling of axons, we focused this study on axons. Axonal labeling was examined in mouse sciatic nerve, and immunoblotting showed the cross-link was restricted to neurofilament heavy and medium subunits, which while altering migration, did not indicate larger NF aggregates, indicative of intermolecular cross-links. Examination of mice at various ages showed the extent of modification remaining relatively constant through the life span. These findings demonstrate lipid-cross-linking peroxidation primarily involves lysine-rich neurofilaments and is restricted to intramolecular cross-links.


Subject(s)
Aldehydes/chemistry , Neurofilament Proteins/chemistry , Neurofilament Proteins/metabolism , Sciatic Nerve/metabolism , Animals , Antibodies/immunology , Fluorescence , Lysine/chemistry , Lysine/immunology , Mice , Mice, Inbred Strains , Sciatic Nerve/chemistry , Sciatic Nerve/cytology
2.
J Clin Endocrinol Metab ; 98(4): E727-31, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23463654

ABSTRACT

CONTEXT: Most current knowledge of pancreatic islet pathophysiology in diabetes mellitus has come from animal models. Even though islets from humans are readily available, only a few come from diabetic donors. We had the uncommon opportunity to acquire islets from humans with type 2 diabetes and used it to perform a study not previously done with human or animal islets. OBJECTIVES: Oxidative stress has been proposed as a mechanism for impaired ß-cell function in type 2 diabetes. Lipid peroxides caused by reactive oxygen species are damaging to body tissues. The objective was to determine whether lipid peroxide-protein adducts occur in pancreatic islets of humans with type 2 diabetes. DESIGN: Immunoblots with two antibodies to hydroxynonenal and 2 other antibodies we generated against reactive small aliphatic compounds were used to detect lipid peroxide-protein adducts in islets of patients with type 2 diabetes and controls. RESULTS: The antibodies reacted strongly to ≥5 islet proteins. The major hydroxynonenal adduct in the islets of type 2 diabetes patients was a 52-kDa protein seen with all 4 antibodies that was also seen in islets of nondiabetic humans, rat islets, and insulinoma cells and in mitochondria of various rat tissues. Nano-LC-MS/MS (liquid chromatography-tandem mass spectrometry) and MALDI-TOF (matrix-assisted laser desorption/ionization-time of flight) analysis identified the protein as the ß-chain of the mitochondrial F-ATP synthase, an enzyme responsible for 95% of ATP formed in tissues. CONCLUSIONS: Lipid peroxide-protein adducts occur in ß-cells in the nondiabetic state and in diabetes. Lipid peroxidation is thought to be damaging to tissues. Analogous to various other unhealthy characteristics, the presence in nondiabetic individuals of lipid peroxide-protein adducts does not necessarily indicate they are not detrimental.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Islets of Langerhans/metabolism , Lipid Peroxides/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Animals , Cells, Cultured , Diabetes Mellitus, Type 2/pathology , Female , Humans , Islets of Langerhans/pathology , Kidney/chemistry , Kidney/metabolism , Kidney/pathology , Kidney/ultrastructure , Lipid Peroxidation/physiology , Mitochondria/chemistry , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proton-Translocating ATPases/isolation & purification , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/isolation & purification , Reactive Oxygen Species/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry
3.
J Neurochem ; 78(2): 413-6, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11461977

ABSTRACT

Glutamate transporters are involved in the maintenance of synaptic glutamate concentrations. Because of its potential neurotoxicity, clearance of glutamate from the synaptic cleft may be critical for neuronal survival. Inhibition of glutamate uptake from the synapse has been implicated in several neurodegenerative disorders. In particular, glutamate uptake is inhibited in Alzheimer's disease (AD); however, the mechanism of decreased transporter activity is unknown. Oxidative damage in brain is implicated in models of neurodegeneration, as well as in AD. Glutamate transporters are inhibited by oxidative damage from reactive oxygen species and lipid peroxidation products such as 4-hydroxy-2-nonenal (HNE). Therefore, we have investigated a possible connection between the oxidative damage and the decreased glutamate uptake known to occur in AD brain. Western blots of immunoprecipitated HNE-immunoreactive proteins from the inferior parietal lobule of AD and control brains suggest that HNE is conjugated to GLT-1 to a greater extent in the AD brain. A similar analysis of beta amyloid (Abeta)-treated synaptosomes shows for the first time that Abeta1-42 also increases HNE conjugation to the glutamate transporter. Together, our data provide a possible link between the oxidative damage and neurodegeneration in AD, and supports the role of excitotoxicity in the pathogenesis of this disorder. Furthermore, our data suggests that Abeta may be a possible causative agent in this cascade.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Aldehydes/pharmacology , Amyloid beta-Peptides/pharmacology , Brain/metabolism , Glutamic Acid/metabolism , Neuroglia/metabolism , Parietal Lobe/metabolism , Peptide Fragments/pharmacology , ATP-Binding Cassette Transporters/drug effects , Aged , Amino Acid Transport System X-AG , Animals , Brain/pathology , Cerebral Cortex/metabolism , Cross-Linking Reagents/pharmacology , Female , Humans , Male , Neuroglia/pathology , Organ Size , Oxidation-Reduction , Parietal Lobe/pathology , Rats , Reference Values , Synaptosomes/drug effects , Synaptosomes/metabolism
4.
Biochemistry ; 40(8): 2548-54, 2001 Feb 27.
Article in English | MEDLINE | ID: mdl-11327877

ABSTRACT

Apolipoprotein E (apoE) plays an important role in the response to central nervous system injury. The e4 allele of apoE and amyloid beta-peptide (Abeta) are associated with Alzheimer's disease (AD) and may be central to the pathogenesis of this disorder. Recent studies demonstrate evidence for neurodegeneration and increased lipid peroxidation in transgenic mice lacking apoE (KO). In the current study, synaptosomes were prepared from apoE KO mice to determine the role of apoE in synaptic membrane structure and to determine susceptibility to oxidative damage by Abeta(1-40). ApoE KO mice exhibited structural modifications to lipid and protein components of synaptosomal membranes as determined by electron paramagnetic resonance in conjunction with lipid- and protein- specific spin labels. Incubation with 5 microM Abeta(1-40) resulted in more severe oxidative modifications to proteins and lipids in apoE KO synaptosomes as measured by protein carbonyls, an index of protein oxidation, and TBARs and protein-bound 4-hydroxynonenal (HNE), markers of lipid oxidation. Together, these data support a role for apoE in the modulation of oxidative injury and in the maintenance of synaptic integrity and are discussed with reference to alterations in AD brain.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/pharmacology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Peptide Fragments/pharmacology , Synaptosomes/chemistry , Synaptosomes/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Hydrogen Peroxide/metabolism , Lipid Peroxidation/genetics , Male , Membrane Fluidity/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Synaptosomes/drug effects , Synaptosomes/pathology , Thiobarbituric Acid Reactive Substances/metabolism
5.
J Biol Chem ; 276(32): 30057-63, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11375979

ABSTRACT

Restoration of blood flow to ischemic myocardial tissue results in an increase in the production of oxygen radicals. Highly reactive, free radical species have the potential to damage cellular components. Clearly, maintenance of cellular viability is dependent, in part, on the removal of altered protein. The proteasome is a major intracellular proteolytic system which degrades oxidized and ubiquitinated forms of protein. Utilizing an in vivo rat model, we demonstrate that coronary occlusion/reperfusion resulted in declines in chymotrypsin-like, peptidylglutamyl-peptide hydrolase, and trypsin-like activities of the proteasome as assayed in cytosolic extracts. Analysis of purified 20 S proteasome revealed that declines in peptidase activities were accompanied by oxidative modification of the protein. We provide conclusive evidence that, upon coronary occlusion/reperfusion, the lipid peroxidation product 4-hydroxy-2-nonenal selectively modifies 20 S proteasome alpha-like subunits iota, C3, and an isoform of XAPC7. Occlusion/reperfusion-induced declines in trypsin-like activity were largely preserved upon proteasome purification. In contrast, loss in chymotrypsin-like and peptidylglutamyl-peptide hydrolase activities observed in cytosolic extracts were not evident upon purification. Thus, decreases in proteasome activity are likely due to both direct oxidative modification of the enzyme and inhibition of fluorogenic peptide hydrolysis by endogenous cytosolic inhibitory protein(s) and/or substrate(s). Along with inhibition of the proteasome, increases in cytosolic levels of oxidized and ubiquitinated protein(s) were observed. Taken together, our findings provide insight into potential mechanisms of coronary occlusion/reperfusion-induced proteasome inactivation and cellular consequences of these events.


Subject(s)
Multienzyme Complexes/antagonists & inhibitors , Myocardial Reperfusion , Oxygen/metabolism , Aldehydes/pharmacology , Animals , Blotting, Western , Cysteine Endopeptidases/metabolism , Cytosol/metabolism , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Endopeptidases/chemistry , Free Radicals/metabolism , Lipid Peroxidation , Male , Multienzyme Complexes/metabolism , Peptide Hydrolases/metabolism , Proteasome Endopeptidase Complex , Protein Isoforms , Rats , Rats, Sprague-Dawley , Trypsin/pharmacology , Ubiquitins/metabolism
6.
J Biol Chem ; 276(26): 23357-61, 2001 Jun 29.
Article in English | MEDLINE | ID: mdl-11283020

ABSTRACT

During normal cellular metabolism, mitochondrial electron transport results in the formation of superoxide anion (O(2)) and subsequently hydrogen peroxide (H(2)O(2)). Because H(2)O(2) increases in concentration under certain physiologic and pathophysiologic conditions and can oxidatively modify cellular components, it is critical to understand the response of mitochondria to H(2)O(2). In the present study, treatment of isolated rat heart mitochondria with H(2)O(2) resulted in a decline and subsequent recovery of state 3 NADH-linked respiration. Alterations in NADH levels induced by H(2)O(2) closely paralleled changes in the rate of state 3 respiration. Assessment of electron transport chain complexes and Krebs cycle enzymes revealed that alpha-ketoglutarate dehydrogenase (KGDH), succinate dehydrogenase (SDH), and aconitase were susceptible to H(2)O(2) inactivation. Of particular importance, KGDH and SDH activity returned to control levels, concurrent with the recovery of state 3 respiration. Inactivation is not because of direct interaction of H(2)O(2) with KGDH and SDH. In addition, removal of H(2)O(2) alone is not sufficient for reactivation. Enzyme activity does not recover unless mitochondria remain intact. The sensitivity of KGDH and SDH to H(2)O(2)-mediated inactivation and the reversible nature of inactivation suggest a potential role for H(2)O(2) in the regulation of KGDH and SDH.


Subject(s)
Hydrogen Peroxide/pharmacology , Mitochondria, Heart/drug effects , Oxidants/pharmacology , Animals , Cell Respiration , Citric Acid Cycle/drug effects , Electron Transport/drug effects , Enzyme Activation/drug effects , Ketoglutarate Dehydrogenase Complex/antagonists & inhibitors , Kinetics , Mitochondria, Heart/enzymology , Mitochondria, Heart/physiology , NADP/metabolism , Rats , Rats, Sprague-Dawley , Succinate Dehydrogenase/antagonists & inhibitors
7.
Chem Biol Interact ; 130-132(1-3): 261-73, 2001 Jan 30.
Article in English | MEDLINE | ID: mdl-11306050

ABSTRACT

Toxic medium chain length alkanals, alkenals, and 4-hydroxyalkenals that are generated during lipid peroxidation are potential substrates for aldehyde dehydrogenase (ALDH) isoforms. We have developed transgenic cell lines to examine the potential for either human ALDH1A1 or ALDH3A1 to protect against damage mediated by these toxic aldehydes. Using crude cytosols from stably transfected cell lines, these aldehydes were confirmed to be excellent substrates for ALDH3A1, but were poorly oxidized by ALDH1A1. Expression of ALDH3A1 by stable transfection in V79 cells conferred a high level of protection against growth inhibition by the medium-chain length aldehyde substrates with highest substrate activity, including hexanal, trans-2-hexenal, trans-2-octenal, trans-2-nonenal, and 4-hydroxy-2-nonenal (HNE). This was reflected in a parallel ability of ALDH3A1 to prevent depletion of glutathione by these aldehydes. Expression of hALDH3 completely blocked the potent induction of apoptosis by HNE in both V79 cells and in a RAW 264.7 murine macrophage cell line, consistent with the observed total prevention of HNE-protein adduct formation. Structure-activity studies indicated that the rank order of potency for the contributions of HNE functional groups to toxicity was aldehyde >/=C2=C3 double bond>>C4-hydroxyl group. Oxidation of the aldehyde moiety of HNE to a carboxyl by ALDH3A1 expressed in stably transfected cell lines drastically reduced its potency for growth inhibition and apoptosis induction. In contrast, ALDH1A1 expression provided only moderate protection against trans-2-nonenal (t2NE), and none against the other six-nine carbon aldehydes. Neither ALDH1A1 nor ALDH3A1 conferred any protection against acrolein, acetaldehyde, or chloroacetaldehyde. A small degree of protection against malondialdehyde was afforded by ALDH1A1, but not ALDH3A1. Paradoxically, cells expressing ALDH3A1 were 1.5-fold more sensitive to benzaldehyde toxicity than control V79 cells. These studies demonstrate that expression of class 3 ALDH, but not class 1 ALDH, can be an important determinant of cellular resistance to toxicity mediated by aldehydes of intermediate chain length that are produced during lipid peroxidation.


Subject(s)
Acetaldehyde/analogs & derivatives , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehydes/pharmacokinetics , Aldehydes/toxicity , Isoenzymes/genetics , Isoenzymes/metabolism , Acetaldehyde/toxicity , Acrolein/toxicity , Aldehyde Dehydrogenase 1 Family , Alkylation , Animals , Apoptosis/drug effects , Benzaldehydes/toxicity , Cell Division/drug effects , Cell Line , Cricetinae , Drug Resistance , Humans , Inactivation, Metabolic , Lipid Peroxidation , Proteins/metabolism , Rats , Retinal Dehydrogenase , Transfection
8.
Free Radic Biol Med ; 30(1): 129-38, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11134903

ABSTRACT

Recent studies indicate that sepsis is associated with enhanced generation of several free radical species (nitric oxide, superoxide, hydrogen peroxide) in skeletal muscle. While studies suggest that free radical generation causes uncoupling of oxidative phosphorylation in sepsis, no previous report has examined the role of free radicals in modulating skeletal muscle oxygen consumption during State 3 respiration or inhibiting the electron transport chain in sepsis. The purpose of the present study was to examine the effects of endotoxin-induced sepsis on State 3 diaphragm mitochondrial oxygen utilization and to determine if inhibitors/scavengers of various free radical species would protect against these effects. We also examined mitochondrial protein electrophoretic patterns to determine if observed endotoxin-related physiological derangements were accompanied by overt alterations in protein composition. Studies were performed on: (a) control animals, (b) endotoxin-treated animals, (c) animals given endotoxin plus PEG-SOD, a superoxide scavenger, (d) animals given endotoxin plus L-NAME, a nitric oxide synthase inhibitor, (e) animals given only PEG-SOD or L-NAME, (f) animals given endotoxin plus D-NAME, and (g) animals given endotoxin plus denatured PEG-SOD. We found: (a) no alteration in maximal State 3 mitochondrial oxygen consumption rate at 24 h after endotoxin administration, but (b) a significant reduction in oxygen consumption rate at 48 h after endotoxin, (c) no effect of endotoxin to induce uncoupling of oxidative phosphorylation, (d) either PEG-SOD or L-NAME (but neither denatured PEG-SOD nor D-NAME) prevented endotoxin-mediated reductions in State 3 respiration rates, (e) some mitochondrial proteins underwent tyrosine nitrosylation at 24 h after endotoxin administration, and (f) SDS-page electrophoresis of mitochondria from endotoxin-treated animals revealed a selective depletion of several proteins at 48 h after endotoxin administration (but not at 24 h); (g) administration of L-NAME or PEG-SOD prevented this protein depletion. These data provide the first evidence that endotoxin-induced reductions in State 3 mitochondrial oxygen consumption are free radical-mediated.


Subject(s)
Diaphragm/ultrastructure , Endotoxins , Mitochondria/metabolism , Oxygen Consumption/drug effects , Sepsis/metabolism , Animals , Electrophoresis, Polyacrylamide Gel , Endotoxins/administration & dosage , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Free Radicals , Hydrogen Peroxide/metabolism , Male , Mitochondria/chemistry , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Polyethylene Glycols/pharmacology , Rats , Sepsis/chemically induced , Superoxide Dismutase/pharmacology , Superoxides/metabolism
9.
Mol Pharmacol ; 58(4): 788-94, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10999949

ABSTRACT

4-Hydroxy-2-nonenal (HNE) is a highly reactive lipid aldehyde byproduct of the peroxidation of cellular membranes. The structure of HNE features three functional groups, a C1 aldehyde, a C2==C3 double bond, and a C4- hydroxyl group, each of which may contribute to the toxicity of the compound. In addition, the length of the aliphatic chain may influence toxic potency by altering lipophilicity. Using analogous compounds that lacked one or more of the structural moieties, the role of each of these structural motifs in the cytotoxicity of HNE was examined in a mouse alveolar macrophage cell line (RAW 264.7) by a cell survival and growth assay. The importance of these functional groups in the potency of HNE for induction of apoptosis was also examined. The rank order of effects on toxicity was C1---aldehyde >/= C2==C3 double bond >> C4---hydroxyl, with parallel results in both the survival/growth inhibition and apoptosis induction assays. The chain length also influenced toxicity in a series of alpha,beta-unsaturated alkenyl aldehydes, with increasing chain length yielding increasing toxicity. To confirm the importance of the aldehyde moiety, and to examine the role of metabolic detoxification in cellular defenses against HNE toxicity, a RAW 264.7 cell line overexpressing human aldehyde dehydrogenase-3 (hALDH3) was generated. This cell line exhibited nearly complete protection against HNE-protein adduct formation as well as HNE-induced apoptosis. These results illustrate the comparative significance of key structural features of HNE in relation to its potent toxicity and induction of apoptosis.


Subject(s)
Aldehydes/pharmacology , Apoptosis , Aldehydes/chemistry , Animals , Cell Division/drug effects , Cells, Cultured , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/pharmacology , Humans , Mice , Structure-Activity Relationship , Transfection
10.
FASEB J ; 14(12): 1825-36, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10973932

ABSTRACT

In laboratory rodents, caloric restriction (CR) retards several age-dependent physiological and biochemical changes in skeletal muscle, including increased steady-state levels of oxidative damage to lipids, DNA, and proteins. We used immunogold electron microscopic (EM) techniques with antibodies raised against 4-hydroxy-2-nonenal (HNE) -modified proteins, dinitrophenol, and nitrotyrosine to quantify and localize the age-dependent accrual of oxidative damage in rhesus monkey vastus lateralis skeletal muscle. Using immunogold EM analysis of muscle from rhesus monkeys ranging in age from 2 to 34 years old, a fourfold maximal increase in levels of HNE-modified proteins was observed. Likewise, carbonyl levels increased approximately twofold with aging. Comparing 17- to 23-year-old normally fed to age-matched monkeys subjected to CR for 10 years, levels of HNE-modified proteins, carbonyls, and nitrotyrosine in skeletal muscle from the CR group were significantly less than control group values. Oxidative damage largely localized to myofibrils, with lesser labeling in other subcellular compartments. Accumulation of lipid peroxidation-derived aldehydes, such as malondialdehyde and 4-hydroxy-2-alkenals, and protein carbonyls were measured biochemically and confirmed the morphological data. Our study is the first to quantify morphologically and localize the age-dependent accrual of oxidative damage in mammalian skeletal muscle and to demonstrate that oxidative damage in primates is lowered by CR.


Subject(s)
Energy Intake/physiology , Muscle, Skeletal/physiology , Oxidative Stress/physiology , Animals , Energy Metabolism/physiology , Immunohistochemistry , Macaca mulatta , Male , Muscle, Skeletal/ultrastructure
11.
J Autoimmun ; 15(1): 51-60, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10936028

ABSTRACT

Previous studies documenting the existence of cross-reactivity between the lipoated (but not unlipoated) forms of the inner lipoyl domain (E2L2) of PDC-E2 [the major autoantigen in Primary biliary cirrhosis (PBC)] and trifluoroacetylated (TFA) proteins, led us to hypothesize that PBC may be due to an initial insult with an environmental agent that cross-reacts with TFA. Therefore, we performed a comparative study of the reactivity of rabbit anti-TFA antibody and anti-lipoic acid (LA) antibody against the mitochondrial autoantigens of human PBC and various TFA and LA conjugated proteins. Whereas both anti-TFA and anti-LA reacted with PDC-E2, the wild-type lipoated form of E2L2, OGDC-E2, E3-BP and LA-KLH, neither reacted with BCOADC-E2 or the non-lipoated form of E2L2. Of interest was that while anti-TFA reacted with PDC-E2, TFA-RSA and LA-KLH, it failed to inhibit PDC-E2 enzyme function. In contrast, anti-LA demonstrated cytoplasmic and mitochondrial staining, and inhibited PDC enzyme activity. Hence, although considerable cross reactivity exists between anti-TFA and anti-LA, the molecular nature of the interaction is clearly different. One of 14 PBC sera reacted weakly with TFA-albumin, whereas four of 14 PBC sera reacted with LA-KLH. Immunohistochemically, both anti-TFA and anti-LA antibodies reacted focally with periportal hepatocytes and bile ducts in both PBC and controls. However, anti-LA produced much stronger focalized staining of the bile ducts of diseased liver. This study suggests that while anti-TFA antibody recognizes lipoic acid-linked enzymes and proteins, the epitope recognized differs from that of anti-LA antibody and PBC autoantibodies. It is unlikely that a response to TFA is the triggering event in PBC. Anti-LA antibodies share a higher degree of similarity to PBC sera providing suggestive evidence that anti-LA antibodies or anti-LA like antibodies (mimotopes) may help define the initiator of the autoimmune response.


Subject(s)
Autoantibodies/chemistry , Autoantibodies/metabolism , Fluoroacetates , Liver Cirrhosis, Biliary/immunology , Molecular Mimicry/immunology , Thioctic Acid/immunology , Trifluoroacetic Acid/immunology , Animals , Antigen-Antibody Reactions , Cattle , Cytosol/drug effects , Cytosol/immunology , Cytosol/metabolism , Dihydrolipoyllysine-Residue Acetyltransferase , Enzyme Inhibitors/immunology , Enzyme-Linked Immunosorbent Assay , Halothane/administration & dosage , Haptens/immunology , Hemocyanins/immunology , Humans , Immune Sera/metabolism , Immunoblotting , Immunohistochemistry , Liver Cirrhosis, Biliary/blood , Liver Cirrhosis, Biliary/enzymology , Microsomes, Liver/drug effects , Microsomes, Liver/immunology , Microsomes, Liver/metabolism , Mollusca , Pyruvate Dehydrogenase Complex/antagonists & inhibitors , Pyruvate Dehydrogenase Complex/metabolism , Rats , Serum Albumin/immunology
12.
Prostate ; 44(2): 144-55, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10881024

ABSTRACT

BACKGROUND: The risk for prostate cancer seems to be reduced by certain antioxidant compounds (vitamins E and A, and selenium). METHODS: Antioxidant enzymes and oxidative damage products were localized in normal prostatic epithelium and malignant glands in primary and metastatic prostatic adenocarcinomas, using well-characterized antibodies and immunoperoxidase techniques. RESULTS: Antioxidant enzymes and four markers of oxidative damage were compared in basal and secretory cells of normal prostatic epithelium and prostate adenocarcinoma cells, and each cell type had unique patterns of enzymes and oxidative damage products. One marker of oxidative damage, a fluorophore derived from 4-hydroxy-2-nonenal-lysine adduction, was found in secretory cells of normal but not malignant epithelium, demonstrating a different oxidative metabolism in normal vs. malignant prostate epithelium. Metastatic lesions from primary prostate cancer had higher levels of manganese superoxide dismutase and nuclear oxidative damage products than did primary tumors. CONCLUSIONS: Antioxidant enzymes and oxidative damage products are modulated in metastatic compared to primary prostate cancer.


Subject(s)
Adenocarcinoma/secondary , Antioxidants/analysis , Bone Neoplasms/secondary , Prostate/enzymology , Prostatic Neoplasms/enzymology , Reactive Oxygen Species/metabolism , 8-Hydroxy-2'-Deoxyguanosine , Adenocarcinoma/enzymology , Adenocarcinoma/metabolism , Antibodies, Monoclonal , Catalase/analysis , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/analysis , Glutathione Peroxidase/analysis , Humans , Immunoenzyme Techniques , Lipofuscin/chemistry , Male , Prostate/cytology , Prostate/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Superoxide Dismutase/analysis , Tyrosine/analogs & derivatives , Tyrosine/analysis
14.
Brain Res ; 847(2): 203-10, 1999 Nov 20.
Article in English | MEDLINE | ID: mdl-10575089

ABSTRACT

"beta Amyloid (Abeta)-induced free radical-mediated neurotoxicity" is a leading hypothesis as a cause of Alzheimer's disease (AD). Abeta increased free radical production and lipid peroxidation in PC12 nerve cells, leading to increased 4-hydroxy-2-nonenal (HNE) production and modification of specific mitochondrial target proteins, apoptosis and cell death. Pretreatment of the cells with isolated ginkgolides, the anti-oxidant component of Ginkgo biloba leaves, or vitamin E, prevented the Abeta-induced increase of reactive oxygen species (ROS). Ginkgolides, but not vitamin E, inhibited the Abeta-induced HNE modification of mitochondrial proteins. However, treatment with these anti-oxidants did not rescue the cells from Abeta-induced apoptosis and cell death. These results indicate that free radicals and lipid peroxidation may not mediate Abeta-induced neurotoxicity.


Subject(s)
Amyloid beta-Peptides/pharmacology , Diterpenes , Free Radical Scavengers/pharmacology , Lactones/pharmacology , Neurons/drug effects , Reactive Oxygen Species/metabolism , Vitamin E/pharmacology , Aldehydes/metabolism , Amyloid beta-Peptides/drug effects , Animals , Apoptosis/drug effects , Apoptosis/physiology , Free Radicals/metabolism , Ginkgolides , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Neurons/metabolism , PC12 Cells/drug effects , Rats
15.
Free Radic Biol Med ; 27(5-6): 695-703, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10490290

ABSTRACT

Both polyclonal and monoclonal antibodies to 4-hydroxy-2-nonenal (HNE) protein adducts were used to identify lipid peroxidation products in normal human kidney and in selected human kidney cancers using immunoperoxidase techniques at the light microscopic level and immunogold techniques at the ultrastructural level. HNE protein adducts were detected in most cell types in normal kidney, although in highly variable amounts. All six morphologic types of renal tumors examined showed some staining with antibodies to HNE protein adducts, although the intensity of staining varied considerably depending on tumor type. Renal oncocytoma and the granular cell variant of renal adenocarcinoma both showed greater cytoplasmic staining for HNE protein adducts than the other tumors examined; these tumors both contain high numbers of mitochondria and suggest that mitochondria are a major source of lipid peroxidation products. To test this possibility, immunogold ultrastructural analysis was performed. HNE protein adducts were identified in nuclei and mitochondria in both normal proximal tubule and three types of renal carcinoma examined; these results localize oxidative damage at the subcellular level in both benign and malignant epithelium to nuclei and mitochondria. In conclusion, HNE protein adducts occur in kidneys in both normal and tumor cells, although immunomorphologic analyses suggest less HNE protein adducts in tumor cells.


Subject(s)
Aldehydes/metabolism , Kidney Neoplasms/metabolism , Neoplasm Proteins/metabolism , Aldehydes/chemistry , Humans , Immunoenzyme Techniques , Kidney Neoplasms/ultrastructure , Kidney Tubules/ultrastructure , Lipid Peroxidation , Microscopy, Immunoelectron , Mitochondria/ultrastructure , Neoplasm Proteins/chemistry
16.
J Appl Physiol (1985) ; 87(3): 1123-31, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10484586

ABSTRACT

The purpose of the present study was to determine whether it is possible to alter the development of fatigue and ablate free radical-mediated lipid peroxidation of the diaphragm during loaded breathing by administering oxypurinol, a xanthine oxidase inhibitor. We studied 1) room-air-breathing decerebrate, unanesthetized rats given either saline or oxypurinol (50 mg/kg) and loaded with a large inspiratory resistance until airway pressure had fallen by 50% and 2) unloaded saline- and oxypurinol-treated room-air-breathing control animals. Additional sets of studies were performed with animals breathing 100% oxygen. Animals were killed at the conclusion of loading, and diaphragmatic samples were obtained for determination of thiobarbituric acid-reactive substances and assessment of in vitro force generation. We found that loading of saline-treated animals resulted in significant diaphragmatic fatigue and thiobarbituric acid-reactive substances formation (P < 0.01). Oxypurinol administration, however, failed to increase load trial time, reduce fatigue development, or prevent lipid peroxidation in either room-air-breathing or oxygen-breathing animals. These data suggest that xanthine oxidase-dependent pathways do not generate physiologically significant levels of free radicals during the type of inspiratory resistive loading examined in this study.


Subject(s)
Enzyme Inhibitors/pharmacology , Lipid Peroxidation/drug effects , Oxypurinol/pharmacology , Physical Exertion/physiology , Respiratory Mechanics/physiology , Xanthine Oxidase/antagonists & inhibitors , Animals , Blood Gas Analysis , Decerebrate State/physiopathology , Diaphragm/drug effects , Diaphragm/metabolism , Free Radicals/metabolism , Hypoxanthines/metabolism , Male , Rats , Rats, Sprague-Dawley , Respiratory Mechanics/drug effects , Time Factors
17.
J Appl Physiol (1985) ; 87(2): 776-82, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10444639

ABSTRACT

Free radicals are known to play an important role in modulating the development of respiratory muscle dysfunction during sepsis. Moreover, neutrophil numbers increase in the diaphragm after endotoxin administration. Whether or not superoxide derived from infiltrating white blood cells contributes to muscle dysfunction during sepsis is, however, unknown. The purpose of the present study was to examine the effect of apocynin, an inhibitor of the superoxide-generating neutrophil NADPH complex, on endotoxin-induced diaphragmatic dysfunction. We studied groups of rats given saline, endotoxin, apocynin, or both endotoxin and apocynin. Animals were killed 18 h after injection, a portion of the diaphragm was used to assess force generation, and the remaining diaphragm was used for determination of 4-hydroxynonenal (a marker of lipid peroxidation) and nitrotyrosine levels (a marker of free radical-mediated protein modification). We found that endotoxin reduced diaphragm force generation and that apocynin partially prevented this decrease [e.g., force in response to 20 Hz was 23 +/- 1 (SE), 12 +/- 2, 23 +/- 1, and 19 +/- 1 N/cm(2), respectively, for saline, endotoxin, apocynin, and endotoxin/apocynin groups; P < 0.001]. Apocynin also prevented endotoxin-mediated increases in diaphragm 4-hydroxynonenal and nitrotyrosine levels (P < 0.01). These data suggest that neutrophil-derived free radicals contribute to diaphragmatic dysfunction during sepsis.


Subject(s)
Acetophenones/therapeutic use , Diaphragm/metabolism , Aldehydes/analysis , Animals , Antioxidants/therapeutic use , Diaphragm/drug effects , Endotoxins/pharmacology , Histocytochemistry , Immunoblotting , Kinetics , Lipid Peroxidation/drug effects , Male , Muscle Contraction/drug effects , Neutrophils/metabolism , Rats , Rats, Sprague-Dawley , Tyrosine/analogs & derivatives , Tyrosine/analysis
18.
Free Radic Biol Med ; 26(9-10): 1181-93, 1999 May.
Article in English | MEDLINE | ID: mdl-10381189

ABSTRACT

Intraperitoneal (IP) injection of ferric nitrilotriacetate (Fe-NTA) to rats and mice results in iron-induced free radical injury and cancer in kidneys. We sought to clarify the exact localization of acute oxidative damage in Fe-NTA-induced nephrotoxicity by performing immunogold light and electron microscopic (EM) techniques using an antibody against 4-hydroxy-2-nonenal (HNE)-modified proteins. Biochemical assays were done to provide complementary quantitative data. Renal accumulation of lipid peroxidation-derived aldehydes, such as malondialdehyde (MDA) and 4-hydroxy-2-alkenals (4-HDA), increased in parallel with protein carbonyl content, an indicator of protein oxidation, 30 min after administration of Fe-NTA. Immunogold light microscopy showed that HNE-modified proteins increased at 30 min with positivity localized to proximal tubular cells. Immunogold EM demonstrated that HNE-modified proteins were mainly in the mitochondria and nuclei of the proximal tubular epithelium. The intensity of labeling at both the light and EM levels increased together with levels of biochemically measured lipid peroxidation products and protein carbonyl content. Our data suggest that the mechanism of acute nephrotoxicity of Fe-NTA involves mitochondrial and nuclear oxidative damage, findings that may help to define the mechanisms of iron-induced cell injury.


Subject(s)
Aldehydes/metabolism , Ferric Compounds/toxicity , Kidney/drug effects , Kidney/metabolism , Nitrilotriacetic Acid/analogs & derivatives , Proteins/metabolism , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Free Radicals/metabolism , Kidney/injuries , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/injuries , Kidney Tubules, Proximal/metabolism , Lipid Peroxidation/drug effects , Male , Malondialdehyde/metabolism , Mice , Microscopy, Immunoelectron , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Nitrilotriacetic Acid/toxicity , Oxidation-Reduction , Rats , Rats, Wistar
19.
Proc Natl Acad Sci U S A ; 96(12): 6689-93, 1999 Jun 08.
Article in English | MEDLINE | ID: mdl-10359773

ABSTRACT

We previously reported that cardiac reperfusion results in declines in mitochondrial NADH-linked respiration. The degree of inactivation increased with age and was paralleled by modification of protein by the lipid peroxidation product 4-hydroxy-2-nonenal. To gain insight into potential sites of oxidative damage, the present study was undertaken to identify specific mitochondrial protein(s) inactivated during ischemia and reperfusion and to determine which of these losses in activity are responsible for observed declines in mitochondrial respiration. Using a Langendorff rat heart perfusion protocol, we observed age-dependent inactivation of complex I during ischemia and complex IV and alpha-ketoglutarate dehydrogenase during reperfusion. Although losses in complex I and IV activities were found not to be of sufficient magnitude to cause declines in mitochondrial respiration, an age-related decrease in complex I activity during ischemia may predispose old animals to more severe oxidative damage during reperfusion. It was determined that inactivation of alpha-ketoglutarate dehydrogenase is responsible, in large part, for observed reperfusion-induced declines in NADH-linked respiration. alpha-Ketoglutarate dehydrogenase is highly susceptible to 4-hydroxy-2-nonenal inactivation in vitro. Thus, our results suggest a plausible mechanism for age-dependent, reperfusion-induced declines in mitochondrial function and identify alpha-ketoglutarate dehydrogenase as a likely site of free radical-mediated damage.


Subject(s)
Aging/physiology , Ketoglutarate Dehydrogenase Complex/physiology , Mitochondria, Heart/physiology , Myocardial Reperfusion Injury/enzymology , Animals , Cell Respiration , Male , Myocardium/enzymology , Myocardium/ultrastructure , NAD/metabolism , Rats , Rats, Inbred F344
20.
Circ Res ; 84(9): 1050-8, 1999 May 14.
Article in English | MEDLINE | ID: mdl-10325242

ABSTRACT

Giant cell arteritis (GCA) is an inflammatory vasculopathy in which T cells and macrophages infiltrate the wall of medium and large arteries. Clinical consequences such as blindness and stroke are related to arterial occlusion. Formation of aortic aneurysms may result from necrosis of smooth muscle cells and fragmentation of elastic membranes. The molecular mechanisms of arterial wall injury in GCA are not understood. To identify mechanisms of arterial damage, gene expression in inflamed and unaffected temporal artery specimens was compared by differential display polymerase chain reaction. Genes differentially expressed in arterial lesions included 3 products encoded by the mitochondrial genome. Immunohistochemistry with antibodies specific for a 65-kDa mitochondrial antigen revealed that increased expression of mitochondrial products was characteristic of multinucleated giant cells and of CD68+ macrophages that cluster in the media and at the media-intima junction. 4-Hydroxy-2-nonenal adducts, products of lipid peroxidation, were detected on smooth muscle cells and on tissue infiltrating cells, in close proximity to multinucleated giant cells and CD68+ macrophages. Also, giant cells and macrophages with overexpression of mitochondrial products were able to synthesize metalloproteinase-2. Our data suggest that in the vascular lesions characteristic for GCA, a subset of macrophages has the potential to support several pathways of arterial injury, including the release of reactive oxygen species and the production of metalloproteinase-2. This macrophage subset is topographically defined and is also identified by overexpression of mitochondrial genes. Because these macrophages have a high potential to promote several mechanisms of arterial wall damage, they should be therapeutically targeted to prevent blood vessel destruction.


Subject(s)
Giant Cell Arteritis/pathology , Giant Cell Arteritis/physiopathology , Macrophages/physiology , Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , Elastic Tissue/physiopathology , Gelatinases/biosynthesis , Gene Expression Regulation/physiology , Humans , Macrophages/immunology , Matrix Metalloproteinase 2 , Metalloendopeptidases/biosynthesis , Mitochondria/physiology , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/physiology , Temporal Arteries/pathology , Temporal Arteries/physiopathology , Tissue Distribution , Tunica Media/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...