Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
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
2.
3.
Neuroscience ; 147(3): 674-9, 2007 Jul 13.
Article in English | MEDLINE | ID: mdl-17570602

ABSTRACT

Acrolein, the most reactive of the alpha,beta-unsaturated aldehydes, is endogenously produced by lipid peroxidation, and has been found increased in the brain of patients with Alzheimer's disease. Although it is known that acrolein increases total protein carbonylation and impairs the function of selected proteins, no study has addressed which proteins are selectively carbonylated by this aldehyde. In this study we investigated the effect of increasing concentrations of acrolein (0, 0.005, 0.05, 0.5, 5, 50 microM) on protein carbonylation in gerbil synaptosomes. In addition, we applied proteomics to identify synaptosomal proteins that were selectively carbonylated by 0.5 microM acrolein. Acrolein increased total protein carbonylation in a dose-dependent manner. Proteomic analysis (two-dimensional electrophoresis followed by mass spectrometry) revealed that tropomyosin-3-gamma isoform 2, tropomyosin-5, beta-actin, mitochondrial Tu translation elongation factor (EF-Tu(mt)) and voltage-dependent anion channel (VDAC) were significantly carbonylated by acrolein. Consistent with the proteomics studies that have identified specifically oxidized proteins in Alzheimer's disease (AD) brain, the proteins identified in this study are involved in a wide variety of cellular functions including energy metabolism, neurotransmission, protein synthesis, and cytoskeletal integrity. Our results suggest that acrolein may significantly contribute to oxidative damage in AD brain.


Subject(s)
Acrolein/pharmacology , Protein Carbonylation/drug effects , Synaptosomes/drug effects , Animals , Brain/ultrastructure , Dose-Response Relationship, Drug , Electrophoresis, Gel, Two-Dimensional/methods , Female , Gerbillinae , Male , Mass Spectrometry/methods
4.
Neuroscience ; 132(2): 313-24, 2005.
Article in English | MEDLINE | ID: mdl-15802185

ABSTRACT

Protein oxidation has been shown to result in loss of protein function. There is increasing evidence that protein oxidation plays a role in the pathogenesis of Alzheimer's disease (AD). Amyloid beta-peptide (1-42) [Abeta(1-42)] has been implicated as a mediator of oxidative stress in AD. Additionally, Abeta(1-42) has been shown to induce cholinergic dysfunction when injected into rat brain, a finding consistent with cholinergic deficits documented in AD. In this study, we used proteomic techniques to examine the regional in vivo protein oxidation induced by Abeta(1-42) injected into the nucleus basalis magnocellularis (NBM) of rat brain compared with saline-injected control at 7 days post-injection. In the cortex, we identified glutamine synthetase and tubulin beta chain 15/alpha, while, in the NBM, we identified 14-3-3 zeta and chaperonin 60 (HSP60) as significantly oxidized. Extensive oxidation was detected in the hippocampus where we identified 14-3-3 zeta, beta-synuclein, pyruvate dehydrogenase, glyceraldehyde-3-phosphate dehydrogenase, and phosphoglycerate mutase 1. The results of this study suggest that a single injection of Abeta(1-42) into NBM can have profound effects elsewhere in the brain. The results further suggest that Abeta(1-42)-induced oxidative stress in rat brain mirrors some of those proteins oxidized in AD brain and leads to oxidized proteins, which when inserted into their respective biochemical pathways yields insight into brain dysfunction that can lead to neurodegeneration in AD.


Subject(s)
Amyloid beta-Peptides/pharmacology , Brain/drug effects , Peptide Fragments/pharmacology , Proteins/metabolism , Proteomics/methods , Animals , Blotting, Western/methods , Brain/metabolism , Electrophoresis, Gel, Two-Dimensional/methods , Immunoprecipitation/methods , Male , Mass Spectrometry/methods , Oxidation-Reduction/drug effects , Rats , Rats, Wistar , Sequence Analysis, Protein
5.
Neuroscience ; 126(4): 915-26, 2004.
Article in English | MEDLINE | ID: mdl-15207326

ABSTRACT

The senescence-accelerated mouse (SAM) is a murine model of accelerated senescence that was established using phenotypic selection. The SAMP series includes nine substrains, each of which exhibits characteristic disorders. SAMP8 is known to exhibit age-dependent learning and memory deficits. In our previous study, we reported that brains from 12-month-old SAMP8 have greater protein oxidation, as well as lipid peroxidation, compared with brains from 4-month-old SAMP8 mice. In order to investigate the relation between age-associated oxidative stress on specific protein oxidation and age-related learning and memory deficits in SAMP8, we used proteomics to identify proteins that are expressed differently and/or modified oxidatively in aged SAMP8 brains. We report here that in 12 month SAMP8 mice brains the expressions of neurofilament triplet L protein, lactate dehydrogenase 2 (LDH-2), heat shock protein 86, and alpha-spectrin are significantly decreased, while the expression of triosephosphate isomerase (TPI) is increased compared with 4-month-old SAMP8 brains. We also report that the specific protein carbonyl levels of LDH-2, dihydropyrimidinase-like protein 2, alpha-spectrin and creatine kinase, are significantly increased in the brain of 12-month-old SAMP8 mice when compared with the 4-month-old SAMP8 brain. These findings are discussed in reference to the effect of specific protein oxidation and changes of expression on potential mechanisms of abnormal alterations in metabolism and neurochemicals, as well as to the learning and memory deficits in aged SAMP8 mice.


Subject(s)
Aging/metabolism , Brain/metabolism , Gene Expression Regulation/physiology , Oxidative Stress/physiology , Proteins/metabolism , Aging/genetics , Animals , Blotting, Western/methods , Electrophoresis, Gel, Two-Dimensional/methods , Male , Mass Spectrometry/methods , Mice , Mice, Inbred Strains , Mice, Neurologic Mutants , Proteomics/methods
6.
J Pharmacol Exp Ther ; 298(2): 461-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11454906

ABSTRACT

Previous studies using transgenic mice in which metallothionein (MT) was overexpressed only in the heart have demonstrated that MT protects from oxidative cardiac injury induced by doxorubicin (DOX), an important anticancer agent. MT cardioprotection is associated with its antiapoptotic effect. The present study was undertaken to test the hypothesis that MT suppresses DOX-induced apoptosis through inhibition of mitochondrial cytochrome c release and caspase-3 activation. Primary cultures of cardiomyocytes isolated from the hearts of transgenic neonatal mice and nontransgenic controls were treated with DOX at a clinically relevant concentration (1.0 microM) for varying time periods. Apoptosis was detected in nontransgenic cardiomyocyte cultures by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling and Annexin V-fluorescein isothiocyanate binding. This apoptotic effect was significantly suppressed in the MT-overexpressing transgenic cardiomyocytes. Western blot analysis revealed that DOX caused mitochondrial cytochrome c release. Furthermore, caspase-3 activation was observed. The activation of this apoptotic pathway by DOX was dramatically inhibited in the MT-overexpressing cardiomyocytes. To elucidate the role of reactive oxygen species (ROS) in the activation of the cytochrome c-mediated caspase-3 activation pathway, the intracellular levels of ROS and their localization were detected by fluorescent confocal microscopy. Mitochondrial ROS concentrations were dramatically elevated by DOX in nontransgenic cardiomyocytes. This elevation was completely inhibited almost in the MT-overexpressing cardiomyocytes. Thus, these results demonstrate that MT suppresses DOX-induced apoptosis in cardiomyocytes through, at least in part, inhibition of the cytochrome c-mediated apoptotic pathway.


Subject(s)
Antibiotics, Antineoplastic/antagonists & inhibitors , Caspases/metabolism , Cytochrome c Group/metabolism , Doxorubicin/antagonists & inhibitors , Metallothionein/pharmacology , Mitochondria, Heart/enzymology , Animals , Animals, Newborn , Annexin A5/metabolism , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Caspase 3 , Doxorubicin/pharmacology , Enzyme Activation/drug effects , Fluorescein-5-isothiocyanate/metabolism , In Situ Nick-End Labeling , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , Mice , Mice, Transgenic , Mitochondria, Heart/drug effects , Myocardium/cytology , Reactive Oxygen Species/metabolism
7.
Cell Signal ; 13(5): 335-43, 2001 May.
Article in English | MEDLINE | ID: mdl-11369515

ABSTRACT

The present study examined the role of mitogen-activated protein kinases (MAPKs) and phosphatidylinositol-3 kinase-stimulated Akt (PI-3K/Akt) in the regulation of constitutive human neutrophil apoptosis by bacterial lipopolysaccharide (LPS) and two chemoattractants, fMLP and leukotriene B(4) (LTB(4)). LPS and LTB(4) inhibited apoptosis, while fMLP had no effect. Inhibition of extracellular signal-regulated kinase (ERK) with PD098059 significantly inhibited the anti-apoptotic effect of both LPS and LTB(4), while inhibition of p38 kinase with SB203580 had no effect. Inhibition of PI-3K with wortmannin and LY294002 significantly attenuated the anti-apoptotic effect of LTB(4), but not LPS. LPS, fMLP, and LTB(4) stimulated similar levels of ERK and Akt activation. LTB(4) and LPS inhibited neutrophil apoptosis when added simultaneously with fMLP, and LTB(4) and LPS demonstrated an additive effect. We conclude that the ERK and/or PI-3K/Akt pathways are necessary, but not sufficient, for LPS and LTB(4) to delay apoptosis, but other anti-apoptotic pathways remain to be identified.


Subject(s)
Apoptosis/immunology , MAP Kinase Signaling System/immunology , Mitogen-Activated Protein Kinases/metabolism , Neutrophils/cytology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Androstadienes/pharmacology , Apoptosis/drug effects , Chemotaxis, Leukocyte/physiology , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , Imidazoles/pharmacology , Leukotriene B4/pharmacology , Lipopolysaccharides/pharmacology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Morpholines/pharmacology , N-Formylmethionine Leucyl-Phenylalanine/analogs & derivatives , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/enzymology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Pyridines/pharmacology , Virulence Factors, Bordetella/pharmacology , Wortmannin , p38 Mitogen-Activated Protein Kinases
8.
Am J Physiol Heart Circ Physiol ; 280(5): H2292-9, 2001 May.
Article in English | MEDLINE | ID: mdl-11299233

ABSTRACT

To study possible mechanisms for metallothionein (MT) inhibition of ischemia-reperfusion-induced myocardial injury, cardiomyocytes isolated from MT-overexpressing transgenic neonatal mouse hearts and nontransgenic controls were subjected to 4 h of hypoxia (5% CO2-95% N2, glucose-free modified Tyrode's solution) followed by 1 h of reoxygenation in MEM + 20% fetal bovine serum (FBS) (5% CO2-95% air), and cytochrome c-mediated caspase-3 activation apoptotic pathway was determined. Hypoxia/reoxygenation-induced apoptosis was significantly suppressed in MT-overexpressing cardiomyocytes, as measured by both terminal deoxynucleotidyl transferase-mediated deoxyuridine 5-triphosphate nick-end labeling and annexin V-FITC binding. In association with apoptosis, mitochondrial cytochrome c release, as determined by Western blot, was observed to occur in nontransgenic cardiomyocytes. Correspondingly, caspase-3 was activated as determined by laser confocal microscopic examination with the use of FITC-conjugated antibody against active caspase-3 and by enzymatic assay. The activation of this apoptotic pathway was significantly inhibited in MT-overexpressing cells, as evidenced by both suppression of cytochrome c release and inhibition of caspase-3 activation. The results demonstrate that MT suppresses hypoxia/reoxygenation-induced cardiomyocyte apoptosis through, at least in part, inhibition of cytochrome c-mediated caspase-3 activation.


Subject(s)
Apoptosis/physiology , Metallothionein/genetics , Muscle Fibers, Skeletal/enzymology , Myocardial Reperfusion Injury/metabolism , Myocardium/cytology , Animals , Caspase 3 , Caspases/metabolism , Cells, Cultured , Cytochrome c Group/metabolism , Gene Expression/physiology , Hypoxia/metabolism , Hypoxia/pathology , In Situ Nick-End Labeling , L-Lactate Dehydrogenase/metabolism , Mice , Mice, Transgenic , Mitochondria/enzymology , Muscle Fibers, Skeletal/cytology , Myocardial Reperfusion Injury/pathology
9.
Am J Physiol Heart Circ Physiol ; 280(4): H1434-41, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11247751

ABSTRACT

The characterization of biological processes on the basis of alterations in the cellular proteins, or "proteomic" analysis, is a powerful approach that may be adopted to decipher the signaling mechanisms that underlie various pathophysiological conditions, such as ischemic heart disease. This review represents a prospectus for the implementation of proteomic analyses to delineate the myocardial intracellular signaling events that evoke cardioprotection against ischemic injury. In concert with this, the manifestation of a protective phenotype has recently been shown to involve dynamic modulation of protein kinase C-epsilon (PKC epsilon) signaling complexes (Ping P, Zhang J, Pierce WM Jr, and Bolli R. Circ Res 88: 59--62, 2001). Accordingly, "the signaling module hypothesis" is formulated as a plausible mechanism by which multipurpose stress-activated proteins and signaling kinases may function collectively to facilitate the genesis of cardioprotection.


Subject(s)
Cardiotonic Agents , Heart/physiology , Isoenzymes/metabolism , Models, Cardiovascular , Protein Kinase C/metabolism , Proteome/metabolism , Signal Transduction/physiology , Animals , Heart/physiopathology , Humans , Myocardial Ischemia/physiopathology , Protein Kinase C-epsilon
10.
J Biol Chem ; 276(5): 3517-23, 2001 Feb 02.
Article in English | MEDLINE | ID: mdl-11042204

ABSTRACT

Akt activation requires phosphorylation of Thr(308) and Ser(473) by 3-phosphoinositide-dependent kinase-1 and 2 (PDK1 and PDK2), respectively. While PDK1 has been cloned and sequenced, PDK2 has yet to be identified. The present study shows that phosphatidylinositol 3-kinase-dependent p38 kinase activation regulates Akt phosphorylation and activity in human neutrophils. Inhibition of p38 kinase activity with SB203580 inhibited Akt Ser(473) phosphorylation following neutrophil stimulation with formyl-methionyl-leucyl-phenylalanine, FcgammaR cross-linking, or phosphatidylinositol 3,4,5-trisphosphate. Concentration inhibition studies showed that Ser(473) phosphorylation was inhibited by 0.3 microm SB203580, while inhibition of Thr(308) phosphorylation required 10 microm SB203580. Transient transfection of HEK293 cells with adenoviruses containing constitutively active MKK3 or MKK6 resulted in activation of both p38 kinase and Akt. Immunoprecipitation and glutathione S-transferase (GST) pull-down studies showed that Akt was associated with p38 kinase, MK2, and Hsp27 in neutrophils, and Hsp27 dissociated from the complex upon activation. Active recombinant MK2 phosphorylated recombinant Akt and Akt in anti-Akt, anti-MK2, anti-p38, and anti-Hsp27 immunoprecipitates, and this was inhibited by an MK2 inhibitory peptide. We conclude that Akt exists in a signaling complex containing p38 kinase, MK2, and Hsp27 and that p38-dependent MK2 activation functions as PDK2 in human neutrophils.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Neutrophils/metabolism , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/pharmacology , Intracellular Signaling Peptides and Proteins , MAP Kinase Kinase 3 , MAP Kinase Kinase 6 , Mitogen-Activated Protein Kinase Kinases/metabolism , Neutrophils/enzymology , Phosphatidylinositols/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-akt , Pyridines/pharmacology , Serine/genetics , Threonine/genetics , p38 Mitogen-Activated Protein Kinases
11.
J Biol Chem ; 275(50): 38957-60, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11042194

ABSTRACT

Previous studies have demonstrated that metallothionein functions as an antioxidant that protects against oxidative DNA, protein, and lipid damage induced by superoxide anion, hydrogen peroxide, hydroxyl radical, and nitric oxide. The present study was undertaken to test the hypothesis that metallothionein also protects from DNA and lipoprotein damage induced by peroxynitrite, an important reactive nitrogen species that causes a diversity of pathological processes. A cell-free system was used. DNA damage was detected by the mobility of plasmid DNA in electrophoresis. Oxidation of low density lipoprotein was measured by a thiobarbituric acid-reactive substance, which was confirmed by lipid hydroperoxide assay. Plasmid DNA damage and low density lipoprotein oxidation were induced by 3-morpholinosydnomine, which produces peroxynitrite through the reaction between nitric oxide and superoxide anion or by synthesized peroxynitrite directly. DNA damage by 3-morpholinosydnomine was prevented by both metallothionein and superoxide dismutase, whereas the damage caused by peroxynitrite was prevented by metallothionein only. The oxidation of low density lipoprotein by 3-morpholinosydnomine and peroxynitrite was also significantly inhibited by metallothionein. This study thus demonstrates that metallothionein may react directly with peroxynitrite to prevent DNA and lipoprotein damage induced by this pathological reactive nitrogen species.


Subject(s)
DNA Damage/drug effects , Lipoproteins/metabolism , Metallothionein/pharmacology , Nitrates/metabolism , Antioxidants/pharmacology , Cell-Free System , Dose-Response Relationship, Drug , Lipid Peroxides/metabolism , Lipoproteins, LDL/metabolism , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , Nitrogen/metabolism , Oxygen/metabolism , Plasmids/metabolism , Temperature , Thiobarbituric Acid Reactive Substances/metabolism , Time Factors
12.
J Leukoc Biol ; 68(2): 277-83, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10947073

ABSTRACT

We investigated intracellular signaling events involved in fibronectin-accelerated TNF-alpha-mediated PMN apoptosis by means of 2-D gel electrophoresis and western blotting. Proteins were sequenced with electrospray ionization mass spectrometry. Apoptosis was quantitated by flow cytometry. We detected a cluster of acidic, high molecular-weight proteins that were only tyrosine phosphorylated when TNF-alpha-treated PMN interacted with fibronectin. Sequence analysis revealed that one of these proteins was Ly-GDI, a regulator of Rho GTPases. Fibronectin increased the TNF-alpha-induced Ly-GDI cleavage, yielding a 23-kD fragment. At 8 h, intact Ly-GDI was decreased to 33% on fibronectin, compared with 69% on PolyHema (P<0.05). Inhibition of tyrosine phosphorylation prevented phosphorylation of Ly-GDI, fibronectin-accelerated Ly-GDI cleavage, and fibronectin-accelerated apoptosis in TNF-alpha-treated PMN. We found that Ly-GDI cleavage was dependent on caspase-3 activation and that caspase-3 inhibition decreased apoptosis. We conclude that tyrosine phosphorylation of Ly-GDI, followed by increased caspase-3-mediated Ly-GDI cleavage, is a signaling event associated with accelerated TNF-alpha-mediated apoptosis on fibronectin.


Subject(s)
Apoptosis/drug effects , Apoptosis/physiology , Neutrophils/pathology , Neutrophils/physiology , Proteins/physiology , Tumor Necrosis Factor-alpha/pharmacology , Cells, Cultured , Guanine Nucleotide Dissociation Inhibitors , Humans , Signal Transduction/drug effects , Signal Transduction/physiology , Tumor Suppressor Proteins , rho GTP-Binding Proteins/physiology , rho Guanine Nucleotide Dissociation Inhibitor beta , rho-Specific Guanine Nucleotide Dissociation Inhibitors
13.
Lab Invest ; 80(5): 745-57, 2000 May.
Article in English | MEDLINE | ID: mdl-10830785

ABSTRACT

Dietary copper restriction causes heart hypertrophy in animal models. Several studies have indicated that this cardiomyopathy is mediated by oxidative stress. Metallothionein (MT), a low molecular weight and cysteine-rich protein, functions in protecting the heart from oxidative injury. We therefore used a cardiac-specific MT-overexpressing transgenic mouse model to test the hypothesis that MT inhibits copper deficiency-induced heart hypertrophy. Dams of both transgenic pups and non-transgenic littermates were fed a copper-adequate or copper-deficient diet, starting on the fourth day post-delivery, and the weanling mice were continued on the dams' diets until they were killed. Heart hypertrophy developed in copper-deficient pups by the fourth week of the combined pre- and post-weaning feeding and aggressively progressed until the end of the experiment (6 weeks). MT overexpression did not prevent the occurrence of heart hypertrophy, but inhibited the progression of this cardiomyopathy, which correlated with its suppression of cardiac lipid peroxidation. Corresponding to the progression of heart hypertrophy, myocardial apoptosis and atrial natriuretic peptide (ANP) production in the left ventricle were detected in non-transgenic copper-deficient mice; these effects were significantly suppressed in transgenic copper-deficient mice. Measurement of apoptosis by TUNEL assay and Annexin V-FITC confocal microscopy in primary cultures of cardiomyocytes revealed that ANP was largely responsible for the myocyte apoptosis and that MT inhibited ANP-induced apoptosis. The data clearly demonstrate that elevation of MT in the heart inhibits oxidative injury and suppresses the progression of heart hypertrophy in copper deficiency, although it does not block its initiation. The results suggest that MT inhibits the transition from heart hypertrophy to failure by suppressing apoptosis through inhibition of both cardiac ANP production and its apoptotic effect.


Subject(s)
Apoptosis , Atrial Natriuretic Factor/physiology , Cardiomegaly/etiology , Copper/deficiency , Metallothionein/pharmacology , Myocardium/pathology , Animals , Antioxidants/pharmacology , Atrial Natriuretic Factor/genetics , Copper/metabolism , In Situ Nick-End Labeling , Mice , Mice, Transgenic , RNA, Messenger/analysis
14.
J Immunol ; 164(12): 6530-7, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10843711

ABSTRACT

Fc gamma Rs mediate immune complex-induced tissue injury. The hypothesis that Fc gamma RIIa and Fc gamma RIIIb control neutrophil responses by activating mitogen-activated protein kinases was examined. Homotypic and heterotypic cross-linking of Fc gamma RIIa and/or Fc gamma RIIIb resulted in a rapid, transient increase in ERK and p38 activity, with maximal stimulation between 1 and 3 min. Fc gamma RIIa and Fc gamma RIIIb stimulated distinct patterns of ERK and p38 activity, and heterotypic cross-linking failed to stimulate synergistic activation of either ERK or p38 activity. Both Fc gamma RIIa and Fc gamma RIIIb required activation of a nonreceptor tyrosine kinase and phosphatidylinositol 3-kinase for stimulation of ERK and p38. Inhibition of ERK activation with PD98059 enhanced H2O2 production stimulated by homotypic and heterotypic Fc gamma R cross-linking. Inhibition of p38 with SB203580 attenuated H2O2 production stimulated by Fc gamma RIIIb or heterotypic cross-linking, but had no effect on Fc gamma RIIa-stimulated H2O2 production. On the other hand, PD98059 inhibited actin polymerization stimulated by Fc gamma R cross-linking, while SB203580 had no effect. Inhibition of actin polymerization with cytochalasin D enhanced p38 activity stimulated by either Fc gamma RIIa or Fc gamma RIIIb, but cytochalasin D only enhanced H2O2 production stimulated by Fc gamma RIIIb. Our data indicate that Fc gamma RIIa and Fc gamma RIIIb independently activate ERK and p38. The two receptors demonstrate different efficacies for ERK and p38 activation, and they do not act cooperatively. ERK and p38 provide stimulatory and inhibitory signals for neutrophil responses to immune complexes. In addition, these data indicate that actin reorganization may play a role in mediating p38-dependent activation of respiratory burst upon stimulation of Fc gamma RIIIb in neutrophils.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Neutrophil Activation/immunology , Neutrophils/enzymology , Neutrophils/immunology , Receptors, IgG/physiology , Actins/metabolism , Actins/physiology , Enzyme Activation/immunology , Humans , Immunophenotyping , Mitogen-Activated Protein Kinases/physiology , Neutrophils/metabolism , Phosphatidylinositol 3-Kinases/physiology , Respiratory Burst/immunology
15.
J Biol Chem ; 275(18): 13690-8, 2000 May 05.
Article in English | MEDLINE | ID: mdl-10788488

ABSTRACT

Cardiomyopathy induced by doxorubicin (DOX) has long been a major impediment of clinical applications of this effective anticancer agent. Previous studies have shown that cardiac-specific metallothionein (MT)-overexpressing transgenic mice are highly resistant to DOX-induced cardiotoxicity. To investigate cellular and molecular mechanisms by which MT participates in this cytoprotection, transgenic mice containing high levels of cardiac MT and non-transgenic controls were treated intraperitoneally with DOX at a single dose of 15 mg/kg and sacrificed on the 4th day after treatment. Myocardial apoptosis was detected by a terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling assay and confirmed by electron microscopy of immunogold staining of apoptotic nuclei. Dual staining of cardiac alpha-sarcomeric actin using an immunohistochemical method further identified apoptotic myocytes. Apoptosis was significantly inhibited in the transgenic myocardium. The anti-apoptotic effect of MT was further revealed in primary cultures of neonatal mouse cardiomyocytes. Furthermore, DOX activated p38 mitogen-activated protein kinase (MAPK), which was critically involved in the apoptotic process, as demonstrated by inhibition of DOX-induced apoptosis by a p38-specific inhibitor, SB203580. Both DOX-induced p38 MAPK activation and apoptosis were dramatically inhibited in the transgenic cardiomyocytes. The results thus demonstrate that DOX induces apoptosis in cardiomyocytes both in vivo and in vitro and MT suppresses this effect through at least in part inhibition of p38 MAPK activation.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Doxorubicin/pharmacology , Metallothionein/pharmacology , Myocardium/metabolism , Myocardium/pathology , Animals , Cells, Cultured , Drug Antagonism , Mice , Signal Transduction/drug effects
16.
J Surg Res ; 89(1): 53-9, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10720453

ABSTRACT

BACKGROUND: Heparin-binding protein (HBP), a serine protease without any known proteolytic activity, is found in human polymorphonuclear leukocyte (PMN) granules, but not in mice. HBP potentiates the endotoxin-induced release of tumor necrosis factor (TNF) alpha, interleukin (IL)-1, and IL-6 from isolated monocytes. HBP has also been shown to increase the survival of cultured monocytes and protect them from oxidative stress. However, whether HBP affects PMNs themselves is not known. MATERIALS AND METHODS: Based on our work with cultured monocytes and the survival benefit noted in experimental peritonitis, we hypothesized that HBP would have a beneficial effect on the survival of neutrophils. We evaluated the effect of HBP on apoptosis in murine peritoneal exudative cells elicited by intraperitoneal thioglycollate administration and in normal human neutrophils from volunteers. Leukocytes were isolated from the peritoneal cavity and blood of mice that underwent intraperitoneal thioglycollate instillation. The mouse peritoneal exudate cells and normal human neutrophils isolated from peripheral blood were used to study the effect of HBP on survival and apoptosis. RESULTS: HBP decreased percentage apoptosis of mouse cells in both serum-enriched (from 24.8 to 4.5%) and serum-deprived (from 23.1 to 8.2%) cultures. In human PMNs, the protective effect of HBP was seen only in the serum-deprived group, with a decrease in percentage apoptosis from 69.1 to 43.3%. CONCLUSIONS: For the first time, we have shown that HBP, in addition to its known augmentation of the proinflammatory response of monocytes, also acts as a prosurvival protein for neutrophils themselves, and thereby enhances local host defense.


Subject(s)
Apoptosis/drug effects , Carrier Proteins/pharmacology , Glycoproteins/pharmacology , Neutrophils/physiology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Humans , LDL-Receptor Related Protein-Associated Protein , Leukocytes/physiology , Mice , Microscopy, Confocal , Neutrophils/drug effects , Peritoneum/cytology , Peritoneum/physiology , Peritonitis/chemically induced , Peritonitis/pathology , Peritonitis/physiopathology , Thioglycolates , Time Factors
17.
J Immunol ; 164(8): 4286-91, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10754327

ABSTRACT

Activated neutrophils play an important role in the pathogenesis of sepsis, glomerulonephritis, acute renal failure, and other inflammatory processes. The resolution of neutrophil-induced inflammation relies, in large part, on removal of apoptotic neutrophils. Neutrophils are constitutively committed to apoptosis, but inflammatory mediators, such as GM-CSF, slow neutrophil apoptosis by incompletely understood mechanisms. We addressed the hypothesis that GM-CSF delays neutrophil apoptosis by activation of extracellular signal-regulated kinase (ERK) and phosphoinositide 3-kinase (PI 3-kinase) pathways. GM-CSF (20 ng/ml) significantly inhibited neutrophil apoptosis (GM-CSF, 32 vs 65% of cells p < 0. 0001). GM-CSF activated the PI 3-kinase/Akt pathway as determined by phosphorylation of Akt and BAD. GM-CSF-dependent Akt and BAD phosphorylation was blocked by the PI 3-kinase inhibitor LY294002. A role for the PI 3-kinase/Akt pathway in GM-CSF-stimulated delay of apoptosis was indicated by the ability of LY294002 to attenuate apoptosis delay. GM-CSF-dependent inhibition of apoptosis was significantly attenuated by PD98059, an ERK pathway inhibitor. LY294002 and PD98059 did not produce additive inhibition of apoptosis delay. To determine whether PI 3-kinase and ERK are used by other ligands that delay neutrophil apoptosis, we examined the role of these pathways in IL-8-induced apoptosis delay. LY294002 blocked IL-8-dependent Akt phosphorylation. PD98059 and LY294002 significantly attenuated IL-8 delay of apoptosis. These results indicate IL-8 and GM-CSF act, in part, to delay neutrophil apoptosis by stimulating PI 3-kinase and ERK-dependent pathways.


Subject(s)
Apoptosis , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Mitogen-Activated Protein Kinases/physiology , Neutrophils/cytology , Neutrophils/enzymology , Phosphatidylinositol 3-Kinases/physiology , Protein Serine-Threonine Kinases , Signal Transduction , Apoptosis/drug effects , Carrier Proteins/metabolism , Cells, Cultured , Chromones/pharmacology , Drug Synergism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Interleukin-8/physiology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Morpholines/pharmacology , Neutrophils/drug effects , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Recombinant Proteins , Signal Transduction/drug effects , Time Factors , bcl-Associated Death Protein
18.
Am J Kidney Dis ; 33(3): 483-91, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10070912

ABSTRACT

The oxidative burst of neutrophils from patients with renal failure before the initiation of dialysis is primed for an enhanced response after stimulation by phagocytosis or chemoattractants. This study shows that phagocytosis-stimulated oxidative burst activity remains primed in patients treated with both high-efficiency hemodialysis and continuous ambulatory peritoneal dialysis (CAPD), but it is normal in patients with a functioning renal transplant. Incubation of normal neutrophils or HL-60 granulocytes in azotemic plasma results in increased resting and phagocytosis-stimulated H2O2 production, which is rapidly reversible on removal of the plasma. Priming of the oxidative burst by azotemic plasma is independent of changes in opsonization and phagocytosis and does not require protein synthesis. These results suggest that azotemic plasma contains a substance or substances capable of reversibly priming oxidative burst activity in neutrophils and neutrophil-like cell lines. The Inability of high-efficiency hemodialysis and CAPD to normalize oxidative burst activity suggests that this substance is of higher molecular weight.


Subject(s)
Kidney Transplantation , Neutrophils/metabolism , Renal Dialysis , Respiratory Burst , Uremia/blood , Uremia/surgery , Adult , Aged , Female , Granulocytes/metabolism , Humans , Hydrogen Peroxide/metabolism , Male , Middle Aged , Peritoneal Dialysis, Continuous Ambulatory , Phagocytosis , Uremia/metabolism
19.
Kidney Int ; 55(2): 562-71, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9987080

ABSTRACT

BACKGROUND: During inflammation, polymorphonuclear neutrophils (PMNs) migrate into the affected tissue interacting with extracellular matrix (ECM) proteins. We tested the hypothesis that PMN-matrix interaction affects PMN apoptosis. METHODS: Apoptosis of human PMNs was detected by DNA-fragmentation assay and was quantitated by flow cytometry, ultraviolet and light microscopy. Cell adhesion was assessed by a toluidine blue assay, and cell spreading was detected by phase contrast microscopy. Protein tyrosine phosphorylation was studied using Western blotting and confocal microscopy. RESULTS: PMN apoptosis was not different in unstimulated cultures on either surface-adherent fibronectin or on PolyHema, a surface that prevents cell adherence. However, tumor necrosis factor-alpha (TNF alpha) treatment significantly increased apoptosis on fibronectin (37 +/- 4%) compared with PolyHema (20 +/- 3%). Tests on other matrix substances revealed that the percentage of apoptotic PMNs in the presence of TNF alpha was 8 +/- 1% on PolyHema, 26 +/- 4% on fibronectin, 17 +/- 2% on collagen I, 16 +/- 2% on collagen IV, and 16 +/- 3% on laminin (P < 0.05 for all matrices compared with PolyHema). Preincubation with genistein (50 microM) significantly inhibited TNF alpha-mediated apoptosis on fibronectin (39 +/- 4% to 21 +/- 4%) but not on PolyHema (21 +/- 4% to 16 +/- 4%). Genistein also reduced PMN spreading on fibronectin. In contrast, inhibitors of mitogen-activated protein kinase and protein kinase C showed no effect on PMN apoptosis. Fibronectin strongly increased tyrosine phosphorylation of three 102, 63, and 54 kDa proteins. Five newly tyrosine-phosphorylated 185, 85, 66, 56, and 42 kDa bands were also visible. Using confocal microscopy, highest tyrosine phosphorylation was localized to sites of cell-matrix interaction. CONCLUSIONS: ECM influences apoptosis in TNF alpha-activated, adherent, spreading PMNs. The process is regulated by tyrosine phosphorylation. Acceleration of apoptosis may shorten the PMN lifespan and thereby locally regulate inflammation.


Subject(s)
Apoptosis/physiology , Extracellular Matrix/physiology , Neutrophils/physiology , Cell Adhesion/drug effects , Fibronectins/physiology , Genistein/pharmacology , Humans , Neutrophils/drug effects , Phosphoproteins/metabolism , Phosphorylation , Polyhydroxyethyl Methacrylate , Time Factors , Tissue Distribution/physiology , Tumor Necrosis Factor-alpha/pharmacology , Tyrosine/metabolism
20.
J Leukoc Biol ; 64(6): 835-44, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9850168

ABSTRACT

The hypothesis that bacterial phagocytosis by human polymorphonuclear neutrophils (PMNs) stimulates MAPK cascades that regulate respiratory burst activation was tested. Extracellular response kinase (ERK) and p38 kinase, but not c-Jun NH2-terminal kinase, activities were increased within 5 min of phagocytosis of plasma-opsonized Staphylococcus aureus (S-SA), reached maximum at 20-30 min, and remained elevated through 60 min. The role of Fcy receptors was examined using gamma globulin-opsonized SA (IgG-SA), whereas CR3 receptors were activated by particulate beta-glucan. IgG-SA stimulated a maximal ERK activity at 30 min, whereas p38 activity was maximal at 5 min. Beta-glucan stimulated maximal ERK activity at 5 min and maximal p38 activity at 2 min. Non-opsonized bacteria were ingested at 10% of the level of S-SA and stimulated a minimal increase in ERK and p38 activity at 60 min. S-SA stimulation of ERK was inhibited by wortmannin, LY294002, and genistein, but not calphostin C; whereas p38 stimulation was inhibited by calphostin C and genistein, but not wortmannin and LY294002. Simultaneous measurement of phagocytosis and H2O2 production by flow cytometry was used to assess the role of ERKs and p38 kinase in phagocytosis. The MEK inhibitor PD098059 had no significant effect on phagocytosis or H2O2 production. The p38 kinase inhibitor SB203580 significantly attenuated H2O2 production, whereas phagocytosis was unaffected. In conclusion, bacterial phagocytosis stimulates ERK and p38 activation by distinct signal transduction pathways. Phagocytosis-stimulated p38 kinase activity is necessary for optimal H2O2 production.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Neutrophils/enzymology , Neutrophils/microbiology , Phagocytosis/physiology , Signal Transduction/physiology , Staphylococcus aureus/physiology , Antigens, CD/physiology , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Enzyme Activation , Humans , JNK Mitogen-Activated Protein Kinases , Macrophage-1 Antigen/physiology , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/physiology , Neutrophils/physiology , Receptors, IgG/physiology , Respiratory Burst/physiology , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...