Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Int J Dev Neurosci ; 70: 1-2, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30053415
3.
J Neurosci Res ; 93(6): 938-47, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25677400

ABSTRACT

Accumulating evidence points to roles for oxidative stress, amyloid beta (Aß), and mitochondrial dysfunction in the pathogenesis of Alzheimer's disease (AD). In neurons, the base excision repair pathway is the predominant DNA repair (BER) pathway for repairing oxidized base lesions. Apurinic/apyrimidinic endonuclease 1 (APE1), a multifunctional enzyme with DNA repair and reduction-oxidation activities, has been shown to enhance neuronal survival after oxidative stress. This study seeks to determine 1) the effect of Aß25-35 on reactive oxygen species (ROS)/reactive nitrogen species (RNS) levels, 2) the activities of respiratory complexes (I, III, and IV), 3) the role of APE1 by ectopic expression, and 4) the neuromodulatory role of ginkgolide B (GB; from the leaves of Ginkgo biloba). The pro-oxidant Aß25-35 peptide treatment increased the levels of ROS/RNS in human neuroblastoma IMR-32 and SH-SY5Y cells, which were decreased after pretreatment with GB. Furthermore, the mitochondrial APE1 level was found to be decreased after treatment with Aß25-35 up to 48 hr, and the level was increased significantly in cells pretreated with GB. The oxidative phosphorylation (OXPHOS; activities of complexes I, III, and IV) indicated that Aß25-35 treatment decreased activities of complexes I and IV, and pretreatment with GB and ectopic APE1 expression enhanced these activities significantly compared with Aß25-35 treatment. Our results indicate that ectopic expression of APE1 potentiates neuronal cells to overcome the oxidative damage caused by Aß25-35 . In addition, GB has been shown to modulate the mitochondrial OXPHOS against Aß25-35 -induced oxidative stress and also to regulate the levels of ROS/RNS in the presence of ectopic APE1. This study presents findings from a new point of view to improve therapeutic potential for AD via the synergistic neuroprotective role played by APE1 in combination with the phytochemical GB.


Subject(s)
Amyloid beta-Peptides/toxicity , Deoxyribonucleases, Type II Site-Specific/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Ginkgolides/pharmacology , Lactones/pharmacology , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Peptide Fragments/toxicity , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Humans , Multienzyme Complexes/metabolism , Neuroblastoma/pathology , Oxidative Phosphorylation/drug effects , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Transfection
4.
Int J Dev Neurosci ; 33: 33-40, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24287098

ABSTRACT

Animal and human studies show that in-utero exposure to preeclampsia alters fetal programming and results in long-term adverse cardiovascular outcomes in the offspring. Human epidemiologic data also suggest that offspring born to preeclamptic mothers are also at risk of adverse long term neurodevelopmental outcomes. Pravastatin, a hydrophilic lipid-lowering drug with pleiotropic properties, was found to prevent the altered cardiovascular phenotype of preeclampsia and restore fetal growth in animal models, providing biological plausibility for its use as a preventive agent for preeclampsia. In this study, we used a murine model of preeclampsia based on adenovirus over-expression of the anti-angiogenic factor soluble Fms-like tyrosine kinase 1, and demonstrated that adult offspring born to preeclamptic dams perform poorly on assays testing vestibular function, balance, and coordination, and that prenatal pravastatin treatment prevents impairment of fetal programming.


Subject(s)
Anticholesteremic Agents/therapeutic use , Gait Disorders, Neurologic/prevention & control , Pravastatin/therapeutic use , Pre-Eclampsia/physiopathology , Prenatal Exposure Delayed Effects/prevention & control , Analysis of Variance , Animals , Disease Models, Animal , Female , Fetal Development/drug effects , Gait Disorders, Neurologic/etiology , Humans , Male , Mice , Postural Balance/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Psychomotor Disorders/etiology , Psychomotor Disorders/prevention & control , Reflex/drug effects , Sex Factors , Transduction, Genetic , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
5.
J Neurosci Res ; 91(9): 1152-64, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23861070

ABSTRACT

Neonatal hypoxia-ischemia (HI) is a common occurrence in preterm and low-birth-weight infants, and the incidence of low-birth-weight and preterm births is increasing. Characterization of brain injury after HI is of critical importance in developing new treatments that more accurately target the injury. After severe HI, neuronal cells undergo necrosis and secondary apoptosis of the surrounding cells as a result of neuroinflammation. We sought to characterize the biochemical pathways associated with cell death after HI. Bax, a cell death signaling protein, is activated after HI and translocates to the nucleus, endoplasmic reticulum, and mitochondria. The translocation patterns of Bax affect the resultant cell death phenotype (necrotic or apoptotic) observed. Although Bax is known to oligomerize once it is activated, less is known about the factors that control its translocation and oligomerization. We hypothesize that Bax kinase-specific phosphorylation determines its oligomerization and intracellular localization. Using well-established in vivo and in vitro models of neonatal HI, we characterized Bax oligomerization and multiorganelle translocation. We found that HI-dependent phosphorylation of Bax determines its oligomerization status and multiorganelle localization, and, ultimately, the cell death phenotype observed. Understanding the mechanisms of Bax translocation will aid in the rational design of therapeutic strategies that decrease the trauma resulting from HI-associated inflammation.


Subject(s)
Cell Nucleus/metabolism , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Neurons/cytology , bcl-2-Associated X Protein/metabolism , Analysis of Variance , Animals , Animals, Newborn , Cell Count , Disease Models, Animal , Embryo, Mammalian , Endoplasmic Reticulum/metabolism , Female , Gene Expression Regulation, Developmental/physiology , Humans , Male , Mitochondria/metabolism , Neuroblastoma/pathology , Neurons/ultrastructure , Phosphorylation , Pregnancy , Rats , Rats, Wistar , Subcellular Fractions , Threonine/metabolism
6.
J Neurotrauma ; 30(9): 727-40, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23360201

ABSTRACT

Mild traumatic brain injury (mTBI), particularly mild "blast type" injuries resulting from improvised exploding devices and many sport-caused injuries to the brain, result in long-term impairment of cognition and behavior. Our central hypothesis is that there are inflammatory consequences to mTBI that persist over time and, in part, are responsible for resultant pathogenesis and clinical outcomes. We used an adaptation (1 atmosphere pressure) of a well-characterized moderate-to-severe brain lateral fluid percussion (LFP) brain injury rat model. Our mild LFP injury resulted in acute increases in interleukin-1α/ß and tumor necrosis factor alpha levels, macrophage/microglial and astrocytic activation, evidence of heightened cellular stress, and blood-brain barrier (BBB) dysfunction that were evident as early as 3-6 h postinjury. Both glial activation and BBB dysfunction persisted for 18 days postinjury.


Subject(s)
Brain Concussion/pathology , Inflammation/pathology , Animals , Blood-Brain Barrier/pathology , Brain/pathology , Brain Concussion/complications , Cytokines/analysis , Cytokines/biosynthesis , Disease Models, Animal , Immunoassay , Inflammation/etiology , Male , Microscopy, Confocal , Motor Activity/physiology , Rats , Rats, Sprague-Dawley
7.
Biochemistry ; 50(25): 5601-14, 2011 Jun 28.
Article in English | MEDLINE | ID: mdl-21615140

ABSTRACT

Cysteinyl S-nitrosylation has emerged as an important post-translational modification affecting protein function in health and disease. Great emphasis has been placed on global, unbiased quantification of S-nitrosylated proteins because of physiologic and oxidative stimuli. However, current strategies have been hampered by sample loss and altered protein electrophoretic mobility. Here, we describe a novel quantitative approach that uses accurate, sensitive fluorescence modification of cysteine S-nitrosylation that leaves electrophoretic mobility unaffected (SNOFlo) and introduce unique concepts for measuring changes in S-nitrosylation status relative to protein abundance. Its efficacy in defining the functional S-nitrosoproteome is demonstrated in two diverse biological applications: an in vivo rat hypoxia-ischemia/reperfusion model and antimicrobial S-nitrosoglutathione-driven transnitrosylation of an enteric microbial pathogen. The suitability of this approach for investigating endogenous S-nitrosylation is further demonstrated using Ingenuity Pathways analysis that identified nervous system and cellular development networks as the top two networks. Functional analysis of differentially S-nitrosylated proteins indicated their involvement in apoptosis, branching morphogenesis of axons, cortical neurons, and sympathetic neurites, neurogenesis, and calcium signaling. Major abundance changes were also observed for fibrillar proteins known to be stress-responsive in neurons and glia. Thus, both examples demonstrate the technique's power in confirming the widespread involvement of S-nitrosylation in hypoxia-ischemia/reperfusion injury and in antimicrobial host responses.


Subject(s)
Cysteine/chemistry , Nitric Oxide/chemistry , Proteomics/methods , Animals , Boron Compounds/chemistry , Calorimetry , Cysteine/metabolism , Female , Fluorescence , Hypoxia/metabolism , Hypoxia/pathology , Ischemia/metabolism , Ischemia/pathology , Luminescence , Maleimides/chemistry , Nitric Oxide/metabolism , Perfusion , Phosphorylation , Random Allocation , Rats , Rats, Wistar
8.
Int J Dev Neurosci ; 29(6): 639-44, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21514373

ABSTRACT

Exposure to hypoxia and hyperoxia in a rodent model of perinatal ischemia results in delayed cell death and inflammation. Hyperoxia increases oxidative stress that can trigger inflammatory cascades, neutrophil activation, and brain microvascular injury. Here we show that 100% oxygen resuscitation in our rodent model of perinatal ischemia increases cortical COX-2 protein levels, S-nitrosylated COX-2cys526, PGE2, iNOS and 5-LOX, all components of the prostaglandin and leukotriene inflammatory pathway.


Subject(s)
Hypoxia-Ischemia, Brain/physiopathology , Oxygen/metabolism , Prostaglandins/metabolism , Animals , Cell Death , Cyclooxygenase 2/metabolism , Female , Humans , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/physiology , Pregnancy , Random Allocation , Rats , Rats, Wistar
9.
J Neurosci Res ; 88(9): 2056-65, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20143414

ABSTRACT

Neonatal hypoxia/ischemia (HI) is a common cause of cognitive and behavioral deficits in children with hyperoxia treatment (HHI) being the current therapy for newborn resuscitation. HI induces cerebral edema that is associated with poor neurological outcomes. Our objective was to characterize cerebral edema after HI and determine the consequences of HHI (40% or 100% O(2)). Dry weight analyses showed cerebral edema 1 to 21 days after HI in the ipsilateral cortex; and 3 to 21 days after HI in the contralateral cortex. Furthermore, HI increased blood-brain barrier (BBB) permeability 1 to 7 days after HI, leading to bilateral cortical vasogenic edema. HHI failed to prevent HI-induced increase in BBB permeability and edema development. At the molecular level, HI increased ipsilateral, but not contralateral, AQP4 cortical levels at 3 and up to 21 days after HI. HHI treatment did not further affect HI-induced changes in AQP4. In addition, we observed developmental increases of AQP4 accompanied by significant reduction in water content and increase permeability of the BBB. Our results suggest that the ipsilateral HI-induced increase in AQP4 may be beneficial and that its absence in the contralateral cortex may account for edema formation after HI. Finally, we showed that HI induced impaired motor coordination 21 days after the insult and HHI did not ameliorate this behavioral outcome. We conclude that HHI treatment is effective as a resuscitating therapy, but does not ameliorate HI-induced cerebral edema and impaired motor coordination.


Subject(s)
Brain Edema/etiology , Brain Edema/therapy , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/therapy , Oxygen Inhalation Therapy/methods , Resuscitation/methods , Animals , Animals, Newborn , Aquaporin 4/metabolism , Blood-Brain Barrier/metabolism , Body Water , Brain/metabolism , Brain Edema/metabolism , Capillary Permeability , Disease Models, Animal , Dyskinesias/complications , Dyskinesias/metabolism , Dyskinesias/therapy , Functional Laterality , Hypoxia-Ischemia, Brain/metabolism , Random Allocation , Rats , Rats, Wistar , Time Factors
10.
Int J Dev Neurosci ; 28(2): 207-16, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19903518

ABSTRACT

Prenatal perturbation of brain circulation and oxygenation is a leading cause of perinatal brain damage affecting about 0.3-0.9% of births. Hypoxia-ischemia (HI) in preterm human infants at gestational week 23-32 results in neurodevelopmental abnormalities in childhood, presenting as learning disability, seizure activity, motor impairment and in the most severe cases, death. Here, we examined the potential of MgSO4 treatment, prior to foetal hypoxia, to attenuate hypoxia induced damage in a murine model of maternal hypoxia. We studied the time course of maternal hypoxia and MgSO4 pre-treatment effects on cerebellar tissue by means of DNA microarray analyses. Mild hypoxia induced minor expression changes in most genes. However, there were 5 gene sets which were down-regulated by maternal hypoxia. MgSO4 pre-treatment abrogated these decreases in gene. A cell cycle gene set which responded immediately (2 h) to hypoxia, showed a delayed response (24 h) when MgSO4 pre-treatment was given. Similar proportions of cell death were observed in all groups before P7, where combined hypoxia and MgSO4 treatment increased cell death in the internal granule layer. There were a higher number of BrdU positive cells at the end of hypoxic episodes and a down-regulation of Reelin signaling, compared to control. MgSO4 pre-treatment prevented the enhancement of cell proliferation due to hypoxia and increased Reelin levels. Altogether, MgSO4 pre-treatment both reduced the number of genes differentially affected by hypoxia and delayed the responses to hypoxia. In addition, MgSO4 pre-treatment modified the nature of the transcriptional response; while hypoxia induced down-regulation of gene sets, MgSO4 pre-treatment mostly up-regulated them. The dual reaction to the MgSO4 treatment may be the source of the ambiguity in observations reported for affected newborns.


Subject(s)
Cerebellum/metabolism , Fetal Hypoxia/drug therapy , Fetal Hypoxia/metabolism , Gene Expression Regulation, Developmental/drug effects , Magnesium Sulfate/administration & dosage , Animals , Dose-Response Relationship, Drug , Mice , Mice, Inbred C57BL , Reelin Protein
11.
Neuropharmacology ; 57(5-6): 511-22, 2009.
Article in English | MEDLINE | ID: mdl-19635490

ABSTRACT

Prenatal hypoxia ischemia is a major cause of neurodevelopmental impairment in the newborn, associated with risk for motor, behavioral and cognitive impaired outcomes. We used an established mouse model of maternal hypoxia to examine the immediate molecular responses of signaling pathways associated with both cell death and neurogenesis. We also characterized responses to maternal pre-treatment with MgSO(4). Maternal hypoxia at embryonic day 17 (E17) failed to trigger inflammation or cell death in fetal brain at 24 h after hypoxia. However, maternal hypoxia decreased levels of neuronal migration signaling: Reelin (53% of control), Disabled 1 (Dab1, 77% of control), and amyloid precursor protein (APP, 64% of control) 2 h after the insult. These changes persisted for 24 h. At later times, Reelin levels in hippocampi of newborns in the maternal hypoxia-treated group increased compared to controls. Full protection from maternal hypoxia effects on hippocampal Reelin levels resulted from maternal pre-treatment with MgSO(4). Hypoxia and MgSO(4) increased radial and lateral migration distance in the CA1 four days after the insult, while in the DG the hypoxia treatment alone increased migration. Maternal hypoxia and MgSO(4) pre-treatment also stimulated hippocampal expression of genes related to neurogenesis, such as BDNF and NeuroD4. Taken together, the long-term neurodevelopmental outcome of prenatal and perinatal hypoxia may depend on perturbation of developmental signals that affect neuronal migration.


Subject(s)
Cell Movement/physiology , Fetal Hypoxia/physiopathology , Hippocampus/embryology , Hippocampus/physiopathology , Hypoxia, Brain/embryology , Hypoxia, Brain/physiopathology , Neurons/physiology , Amyloid beta-Protein Precursor/metabolism , Animals , Animals, Newborn , Cell Adhesion Molecules, Neuronal/metabolism , Cell Death/physiology , Cell Movement/drug effects , Central Nervous System Agents/pharmacology , Dentate Gyrus/drug effects , Dentate Gyrus/embryology , Dentate Gyrus/physiopathology , Disease Models, Animal , Encephalitis/embryology , Encephalitis/physiopathology , Extracellular Matrix Proteins/metabolism , Fetal Hypoxia/drug therapy , Hippocampus/drug effects , Hypoxia, Brain/drug therapy , Magnesium Sulfate/pharmacology , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Neurogenesis/genetics , Neurogenesis/physiology , Neurons/drug effects , Random Allocation , Reelin Protein , Serine Endopeptidases/metabolism , Signal Transduction , Time Factors
12.
Int J Dev Neurosci ; 27(4): 299-304, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19460623

ABSTRACT

During embryonic and early postnatal development the combination of cell proliferation, migration, survival and cell death is intimately regulated. In the mouse embryo, significant numbers of primordial germ cells, the founder cells of the gametes, fail to migrate correctly to the genital ridges early in histogenesis. Studies in Bcl-2 associated X protein null mice (Bax(-/-)) have shown that the pro-apoptotic Bax gene is required for the programmed cell death of germ cells left in ectopic locations during and after germ cell migration. Independent studies carried out in the central nervous system of Bax(-/-) mice have shown impaired and ectopic neuronal migration in the cerebellum and olfactory bulb during development and in the adult hippocampus. Taken together, these evidences identify Bax as a major mechanism in ectopic cell death and are the subject of this review.


Subject(s)
Cell Death/physiology , Embryo, Mammalian/physiology , bcl-2-Associated X Protein/metabolism , Animals , Brain/anatomy & histology , Brain/embryology , Cell Movement/physiology , Embryo, Mammalian/cytology , Germ Cells/cytology , Germ Cells/physiology , Mice , Mice, Knockout , bcl-2-Associated X Protein/genetics
13.
J Neurosci Res ; 87(9): 2047-65, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19224578

ABSTRACT

Neonatal (P7) brain hypoxia-ischemia (HI) induces intracellular Bax protein shifts to the nucleus, mitochondria, and endoplasmic reticulum (ER), where it triggers the activation of the respective cell death signaling cascades. When compared with HI-treated rat pups, 100% O(2) resuscitation of HI-treated rat pups increases HI-induced ER Bax levels, ER-mediated cell death signaling, and resultant lesion volume and inflammation due to increased necrotic-like cell death. To better characterize the role of Bax intracellular shuttling ER cell death signaling and necrotic-like cell death, we used rotenone-treated P5 neuronal cortical cultures to increase ER Bax levels and subsequent cell death signaling. We treated P5 primary cortical neurons with 25 microM and 100 microM rotenone as an apoptotic or necrotic-like stimulus, respectively, and measured intracellular organelle Bax levels and the subsequent activation of ER/mitochondrial cell death signaling. The 25 microM rotenone treatment promptly increased nuclear Bax levels followed by a later increase in mitochondrial Bax levels and caspase-mediated cleavage of alpha-fodrin. The 100 microM rotenone treatment also resulted in an early increase in nuclear Bax levels followed by a subsequent increase in ER Bax levels and calpain-mediated cleavage of alpha-fodrin. After pretreatment with the immunosuppressive and neuroprotective FK506, there was a delay in Bax intracellular shifts and cell death signaling for both the 25 and 100 microM rotenone treatments. These results suggest that the different outcomes of apoptotic-like vs. necrotic-like cell death resulting from the treatment of neuronal cultures with rotenone at 25 and 100 microM rotenone reflect changes in the intracellular trafficking of Bax among different organelles.


Subject(s)
Apoptosis/physiology , Necrosis/metabolism , Neurons/metabolism , Rotenone/pharmacology , bcl-2-Associated X Protein/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Carrier Proteins/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/physiopathology , Microfilament Proteins/metabolism , Necrosis/chemically induced , Necrosis/physiopathology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Organelles/drug effects , Organelles/metabolism , Phenotype , Protein Transport/drug effects , Protein Transport/physiology , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology , Uncoupling Agents/pharmacology
14.
Am J Physiol Heart Circ Physiol ; 295(6): H2436-46, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18931027

ABSTRACT

Endothelial nitric oxide synthase (eNOS) activation with subsequent inducible NOS (iNOS), cytosolic phospholipase A2 (cPLA2), and cyclooxygenase-2 (COX2) activation is essential to statin inhibition of myocardial infarct size (IS). In the rat, the peroxisome proliferator-activated receptor-gamma agonist pioglitazone (Pio) limits IS, upregulates and activates cPLA2 and COX2, and increases myocardial 6-keto-PGF1alpha levels without activating eNOS and iNOS. We asked whether Pio also limits IS in eNOS-/- and iNOS-/- mice. Male C57BL/6 wild-type (WT), eNOS-/-, and iNOS-/- mice received 10 mg.kg(-1).day(-1) Pio (Pio+) or water alone (Pio-) for 3 days. Mice underwent 30 min coronary artery occlusion and 4 h reperfusion, or hearts were harvested and subjected to ELISA and immunoblotting. As a result, Pio reduced IS in the WT (15.4+/-1.4% vs. 39.0+/-1.1%; P<0.001), as well as in the eNOS-/- (32.0+/-1.6% vs. 44.2+/-1.9%; P<0.001) and iNOS-/- (18.0+/-1.2% vs. 45.5+/-2.3%; P<0.001) mice. The protective effect of Pio in eNOS-/- mice was smaller than in the WT (P<0.001) and iNOS-/- (P<0.001) mice. Pio increased myocardial Ser633 and Ser1177 phosphorylated eNOS levels in the WT and iNOS-/- mice. iNOS was undetectable in all six groups. Pio increased cPLA2, COX2, and PGI2 synthase levels in the WT, as well as in the eNOS-/- and iNOS-/-, mice. Pio increased the myocardial 6-keto-PGF1alpha levels and cPLA2 and COX2 activity in the WT, eNOS-/-, and iNOS-/- mice. In conclusion, the myocardial protective effect of Pio is iNOS independent and may be only partially dependent on eNOS. Because eNOS activity decreases with age, diabetes, and advanced atherosclerosis, this effect may be relevant in a clinical setting and should be further characterized.


Subject(s)
Cardiovascular Agents/pharmacology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocardium/enzymology , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type II/deficiency , Thiazolidinediones/pharmacology , 6-Ketoprostaglandin F1 alpha/metabolism , Animals , Cyclooxygenase 2/metabolism , Cytochrome P-450 Enzyme System/metabolism , Disease Models, Animal , Immunoblotting , Intramolecular Oxidoreductases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type III/genetics , Phospholipases A2, Cytosolic/metabolism , Phosphorylation , Pioglitazone , Polymerase Chain Reaction , RNA, Messenger/metabolism
15.
Am J Physiol Heart Circ Physiol ; 295(4): H1809-14, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18676689

ABSTRACT

Decreased cerebral blood flow (CBF) has been observed following the resuscitation from neonatal hypoxic-ischemic injury, but its mechanism is not known. We address the hypothesis that reduced CBF is due to a change in nitric oxide (NO) and superoxide anion O(2)(-) balance secondary to endothelial NO synthase (eNOS) uncoupling with vascular injury. Wistar rats (7 day old) were subjected to cerebral hypoxia-ischemia by unilateral carotid occlusion under isoflurane anesthesia followed by hypoxia with hyperoxic or normoxic resuscitation. Expired CO(2) was determined during the period of hyperoxic or normoxic resuscitation. Laser-Doppler flowmetry was used with isoflurane anesthesia to monitor CBF, and cerebral perivascular NO and O(2)(-) were determined using fluorescent dyes with fluorescence microscopy. The effect of tetrahydrobiopterin supplementation on each of these measurements and the effect of apocynin and N(omega)-nitro-L-arginine methyl ester (L-NAME) administration on NO and O(2)(-) were determined. As a result, CBF in the ischemic cortex declined following the onset of resuscitation with 100% O(2) (hyperoxic resuscitation) but not room air (normoxic resuscitation). Expired CO(2) was decreased at the onset of resuscitation, but recovery was the same in normoxic and hyperoxic resuscitated groups. Perivascular NO-induced fluorescence intensity declined, and O(2)(-)-induced fluorescence increased in the ischemic cortex after hyperoxic resuscitation up to 24 h postischemia. L-NAME treatment reduced O(2)(-) relative to the nonischemic cortex. Apocynin treatment increased NO and reduced O(2)(-) relative to the nonischemic cortex. The administration of tetrahydrobiopterin following the injury increased perivascular NO, reduced perivascular O(2)(-), and increased CBF during hyperoxic resuscitation. These results demonstrate that reduced CBF follows hyperoxic resuscitation but not normoxic resuscitation after neonatal hypoxic-ischemic injury, accompanied by a reduction in perivascular production of NO and an increase in O(2)(-). The finding that tetrahydrobiopterin, apocynin, and L-NAME normalized radical production suggests that the uncoupling of perivascular NOS, probably eNOS, due to acquired relative tetrahydrobiopterin deficiency occurs after neonatal hypoxic-ischemic brain injury. It appears that both NOS uncoupling and the activation of NADPH oxidase participate in the changes of reactive oxygen concentrations seen in cerebral hypoxic-ischemic injury.


Subject(s)
Cerebral Cortex/blood supply , Cerebrovascular Circulation , Endothelium, Vascular/metabolism , Hypoxia-Ischemia, Brain/metabolism , Nitric Oxide/metabolism , Oxygen Inhalation Therapy , Resuscitation , Superoxides/metabolism , Acetophenones/pharmacology , Animals , Animals, Newborn , Biopterins/analogs & derivatives , Biopterins/pharmacology , Carbon Dioxide/metabolism , Cerebrovascular Circulation/drug effects , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Enzyme Inhibitors/pharmacology , Exhalation , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , Hypoxia-Ischemia, Brain/therapy , Laser-Doppler Flowmetry , Microscopy, Fluorescence , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Oxygen Inhalation Therapy/adverse effects , Rats , Rats, Wistar , Resuscitation/adverse effects , Time Factors
16.
J Neurosci Res ; 86(16): 3584-604, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18655197

ABSTRACT

Perinatal hypoxia-ischemia (HI) occurs in 0.2%-0.4% of all live births, with 100% O(2) resuscitation (HHI) remaining a standard clinical treatment. HI produces a broad spectrum of neuronal death phenotypes ranging from a more noninflammatory apoptotic death to a more inflammatory necrotic cell death that may be responsible for the broad spectrum of reported dysfunctional outcomes. However, the mechanisms that would account for this phenotypic spectrum of cell death are not fully understood. Here, we provide evidence that Bcl-2-associated X protein (Bax) can shuttle to different subcellular compartments in response to HI, thus triggering the different organelle-associated cell death signaling cascades resulting in cell death phenotype diversity. There was an early increase in intranuclear and total nuclear Bax protein levels followed by a later Bax redistribution to the mitochondria and endoplasmic reticulum (ER). Associated with the organelle-specific Bax shuttling time course, there was an increase in nuclear phosphorylated p53, cytosolic cleaved caspase-3, and caspase-12. When HI-treated P7 rats were resuscitated with 100% O(2) (HHI), there were increased lesion volumes as determined by T2-weighted magnetic resonance imaging with no change in cortical apoptotic signaling compared with HI treatment alone. There was, however, increased inflammatory (cytosolic-cleaved interleukin-1beta) and necrotic (increased nuclear 55-kDa-cleaved PARP-1 [poly-ADP-ribose 1] and decreased nuclear HMGB1 [nuclear high-mobility group box 1]) after HHI. Furthermore, HHI increased ER calpain activation and ER Bax protein levels compared with HI alone. These data suggest that 100% O(2) resuscitation increases Bax-mediated activation of ER cell death signaling, inflammation, and lesion volume by increasing necrotic-like cell death. In light of these findings, the use of 100% O(2) treatment for neonatal HI should be reevaluated.


Subject(s)
Brain Infarction/metabolism , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/therapy , Nerve Degeneration/metabolism , Oxygen Inhalation Therapy/adverse effects , bcl-2-Associated X Protein/metabolism , Active Transport, Cell Nucleus/physiology , Animals , Animals, Newborn , Apoptosis/physiology , Apoptosis Regulatory Proteins/metabolism , Brain Infarction/etiology , Brain Infarction/physiopathology , Cell Compartmentation/physiology , Cytoplasm/metabolism , Disease Models, Animal , Hyperoxia/etiology , Hyperoxia/metabolism , Hyperoxia/physiopathology , Hypoxia-Ischemia, Brain/physiopathology , Inflammation Mediators/metabolism , Necrosis/etiology , Necrosis/metabolism , Necrosis/physiopathology , Nerve Degeneration/etiology , Nerve Degeneration/physiopathology , Neurons/metabolism , Neurons/pathology , Phenotype , Protein Transport/physiology , Rats , Rats, Wistar , Signal Transduction/physiology
17.
Neurochem Res ; 33(12): 2379-89, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18408999

ABSTRACT

The examination of Bcl-2-associated X protein (Bax) protein's role in the activation of cognate nuclear, mitochondrial and ER cell death signaling cascades and the resulting effects on cell death phenotype in the brain after neonatal hypoxia-ischemia (HI) requires an understanding of neonatal HI insult and progression, as well as, its dysfunctional outcomes. In addition, knowledge of key concepts of oxidative stress, a major injurious component of HI, and the different cell death phenotypes (i.e. apoptosis and necrosis) will aid the design of appropriate useful experimental paradigms. Here we discuss organelle cell death signaling cascades in the context of the different cell death phenotypes associated with animal models of neonatal hypoxia ischemia and tissue culture models used in the study of hypoxia ischemia, focusing on the intracellular shifts of the Bcl-2 associated X protein (Bax) in the hypoxic brain.


Subject(s)
Brain Ischemia/pathology , Brain/pathology , Cell Death , Hypoxia, Brain/pathology , Animals , Animals, Newborn , Oxidative Stress , Rats , Signal Transduction , bcl-2-Associated X Protein/physiology
18.
Exp Neurol ; 210(2): 295-307, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18302959

ABSTRACT

Long-term functional impairments due to spinal cord injury (SCI) in the rat result from secondary apoptotic death regulated, in part, by SCI-induced decreases in protein levels of the antiapoptotic protein Bcl-xL. We have shown that exogenous administration of Bcl-xL spares neurons 24 h after SCI. However, long-term effects of chronic application of Bcl-xL have not been characterized. To counteract SCI-induced decreases in Bcl-xL and resulting apoptosis, we used the TAT protein transduction domain fused to the Bcl-xL protein (Tat-Bcl-xL), or its antiapoptotic domain BH4 (Tat-BH4). We used intrathecal delivery of Tat-Bcl-xL, or Tat-BH4, into injured spinal cords for 24 h or 7 days, and apoptosis, neuronal death and locomotor recovery were assessed up to 2 months after injury. Both, Tat-Bcl-xL and Tat-BH4, significantly decreased SCI-induced apoptosis in thoracic segments containing the site of injury (T10) at 24 h or 7 days after SCI. However, the 7-day delivery of Tat-Bcl-xL, or Tat-BH4, also induced a significant impairment of locomotor recovery that lasted beyond the drug delivery time. We found that the 7-day administration of Tat-Bcl-xL, or Tat-BH4, significantly increased non-apoptotic neuronal loss and robustly augmented microglia/macrophage activation. These results indicate that the antiapoptotic treatment targeting Bcl-xL shifts neuronal apoptosis to necrosis, increases the inflammatory response and impairs locomotor recovery. Our results suggest that a combinatorial treatment consisting of antiapoptotic and anti-inflammatory agents may be necessary to achieve tissue preservation and significant improvement in functional recovery after SCI.


Subject(s)
Apoptosis/physiology , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/therapy , bcl-X Protein/administration & dosage , Analysis of Variance , Animals , Apoptosis/drug effects , Behavior, Animal , CD11b Antigen/metabolism , Disease Models, Animal , Drug Administration Schedule , Enzyme-Linked Immunosorbent Assay/methods , Gene Expression Regulation/drug effects , Gene Products, tat/administration & dosage , Male , Motor Activity/drug effects , Motor Activity/physiology , Protein Structure, Tertiary/physiology , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Recovery of Function/physiology , Spinal Cord Injuries/pathology , Time Factors
19.
J Neurosci Res ; 86(3): 566-80, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-17918744

ABSTRACT

Spinal cord injury (SCI) results in a pathophysiology characterized by multiple locomotor and sensory deficits, resulting in altered nociception and hyperalgesia. SCI triggers an early and prolonged inflammatory response, with increased interleukin-1beta levels. Transient changes are observed in subunit populations of the transcription factor nuclear factor-kappaB (NF-kappaB). There were decreases in neuronal c-Rel levels and inverse increases in p65 and p50 levels. There were no changes in neuronal p52 or RelB subunits after SCI at any time point tested. Similarly, SCI had no effect on oligodendroglial levels of any NF-kappaB subunit. There were significant early increases in COX-2 and inducible nitric oxide synthase mRNA and protein levels after SCI. We used synthetic double-stranded "decoy" deoxyoligonucleotides containing selective NF-kappaB protein dimer binding consensus sequences. Decoys targeting the p65/p50 binding site on the COX-2 promoter decreased SCI-induced cell losses, NF-kappaB p65/p50 DNA-binding activity, and COX-2 and iNOS protein levels. NF-kappaB p65/p50 targeted decoys improved early locomotor recovery after moderate but not severe SCI, yet ameliorated SCI-induced hypersensitization after both moderate and severe SCI. To determine whether changes in GABA activity played a role in decreased hypersensitivity after SCI and p65/p50 targeted decoy, we counted gamma-aminobutyric acid (GABA)-containing neurons in laminae 1-3. There were significantly more GABAergic neurons in the p65/p50 targeted decoy-treated group at the level of injury.


Subject(s)
Behavior, Animal/drug effects , Enzyme Inhibitors/pharmacology , Inflammation/etiology , Inflammation/metabolism , Oligodeoxyribonucleotides/pharmacology , Spinal Cord Injuries/complications , Animals , Binding Sites/drug effects , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , DNA/metabolism , Gene Expression/drug effects , Male , Motor Activity/drug effects , NF-kappa B p50 Subunit/metabolism , Neurons/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Promoter Regions, Genetic , Proto-Oncogene Proteins c-rel/metabolism , Rats , Rats, Sprague-Dawley , Recovery of Function , Spinal Cord Injuries/genetics , Spinal Cord Injuries/physiopathology , Transcription Factor RelA/metabolism , gamma-Aminobutyric Acid/metabolism
20.
Int J Dev Neurosci ; 26(1): 77-85, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17936537

ABSTRACT

The fetal and newborn brain is particularly susceptible to hypoxia, which increases the risk for neurodevelopmental deficits, seizures, epilepsy and life-span motor, behavioral and cognitive disabilities. Here, we report that prenatal hypoxia at gestation day 17 in mice caused an immediate decrease in fetal cerebral cortex levels of glutamate decarboxylase, a key proteins in the GABA pathway. While maternal MgSO4 treatment prior to hypoxia did not have an early effect, it did accelerate maturation at a later stage based on the observed protein expression profile. In addition, MgSO4 reversed the hypoxia-induced loss of a subpopulation of inhibitory neurons that express calbindin in cortex at postnatal day 14. In the hippocampus, responses to prenatal hypoxia were also evident 4 days after the hypoxia. However, in contrast to the observations in cerebral cortex, hypoxia stimulated key protein expression in the hippocampus. The hippocampal response to hypoxia was also reversed by maternal MgSO4 treatment. The data presented here suggests that decreased levels of key proteins in the GABA pathway in the cerebral cortex may lead to high susceptibility to seizures and epilepsy in newborns after prenatal or perinatal hypoxia and that maternal MgSO4 treatment can reverse the hypoxia-induced deficits in the GABA pathway.


Subject(s)
Cerebral Cortex , Down-Regulation/physiology , Hypoxia , Magnesium Sulfate/therapeutic use , Neuroprotective Agents/therapeutic use , Signal Transduction , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Cerebral Cortex/drug effects , Cerebral Cortex/growth & development , Cerebral Cortex/pathology , Down-Regulation/drug effects , Embryo, Mammalian , Female , Glutamate Decarboxylase/metabolism , Hypoxia/drug therapy , Hypoxia/pathology , Hypoxia/physiopathology , Male , Mice , Pregnancy , Signal Transduction/drug effects , Signal Transduction/physiology , Symporters/metabolism , Time Factors , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , K Cl- Cotransporters
SELECTION OF CITATIONS
SEARCH DETAIL
...