Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Toxicol Lett ; 279: 107-114, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28751209

ABSTRACT

Hypoxia is a primary factor in many pathological conditions. Hypoxic cell death is commonly attributed to metabolic failure and oxidative injury. cAMP-dependent protein kinase A (PKA) is activated in hypoxia and regulates multiple enzymes of the mitochondrial electron transport chain, thus may be implicated in cellular energy depletion and hypoxia-induced cell death. Wild type (WT) PC-12 cells and PKA activity-deficient 123.7 PC-12 cells were exposed to 3, 6, 12 and 24h hypoxia (0.1% or 5% O2). Hypoxia, at 24h 0.1% O2, induced cell death and increased reactive oxygen species (ROS) in WT PC-12 cells. Despite lower ATP levels in normoxic 123.7 cells than in WT cells, hypoxia only decreased ATP levels in WT cells. However, menadione-induced oxidative stress similarly affected both cell types. While mitochondrial COX IV expression remained consistently higher in 123.7 cells, hypoxia decreased COX IV expression in both cell types. N-acetyl cysteine antioxidant treatment blocked hypoxia-induced WT cell death without preventing ATP depletion. Transient PKA catα expression in 123.7 cells partially restored hypoxia-induced ROS but did not alter ATP levels or COX IV expression. We conclude that PKA signaling contributes to hypoxic injury, by regulating oxidative stress rather than by depleting ATP levels. Therapeutic strategies targeting PKA signaling may improve cellular adaptation and recovery in hypoxic pathologies.


Subject(s)
Adrenal Gland Neoplasms/enzymology , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Neurons/enzymology , Oxidative Stress , Pheochromocytoma/enzymology , Reactive Oxygen Species/metabolism , Tumor Hypoxia , Adenosine Triphosphate/metabolism , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/pathology , Animals , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , Electron Transport Complex IV/metabolism , Energy Metabolism , Neurons/drug effects , Neurons/pathology , Oxidative Stress/drug effects , PC12 Cells , Pheochromocytoma/genetics , Pheochromocytoma/pathology , Rats , Signal Transduction , Time Factors , Transfection , Vitamin K 3/pharmacology
2.
Life Sci ; 134: 56-62, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26044512

ABSTRACT

Diabetes and its cardiovascular complications have been a major public health issue. These complications are mainly attributable to a severe imbalance between free radical and reactive oxygen species production and the antioxidant defense systems. Nuclear factor E2-related factor 2 (Nrf2) is a transcription factor that controls the basal and inducible expression of a battery of antioxidant enzyme genes and other cyto-protective phase II detoxifying enzymes. As a result, Nrf2 has gained great attention as a promising drug target for preventing diabetic cardiovascular complications. And while animal studies have shown that several Nrf2 activators manifest a potential to efficiently prevent the diabetic complications, their use in humans has not been approved due to the lack of substantial evidence regarding safety and efficacy of the Nrf2 activation. We provide here a brief review of a few clinically-used drugs that can up-regulate Nrf2 with the potential of extending their usage to diabetic patients for the prevention of cardiovascular complications and conclude with a closer inspection of dimethyl fumarate and its mimic members.


Subject(s)
Antioxidants/metabolism , Dermatologic Agents/pharmacology , Diabetic Cardiomyopathies/metabolism , Fumarates/pharmacology , NF-E2-Related Factor 2/biosynthesis , Up-Regulation/drug effects , Animals , Diabetic Cardiomyopathies/drug therapy , Dimethyl Fumarate , Humans
3.
PLoS One ; 9(7): e100230, 2014.
Article in English | MEDLINE | ID: mdl-25014412

ABSTRACT

Hypoxia alters cellular metabolism and although the effects of sustained hypoxia (SH) have been extensively studied, less is known about chronic intermittent hypoxia (IH), commonly associated with cardiovascular morbidity and stroke. We hypothesize that impaired glutamate homeostasis after chronic IH may underlie vulnerability to stroke-induced excitotoxicity. P16 organotypic hippocampal slices, cultured for 7 days were exposed for 7 days to IH (alternating 2 min 5% O2-15 min 21% O2), SH (5% O2) or RA (21% O2), then 3 glutamate challenges. The first and last exposures were intended as a metabolic stimulus (200 µM glutamate, 15 min); the second emulated excitotoxicity (10 mM glutamate, 10 min). GFAP, MAP2, and EAAT1, EAAT2 glutamate transporters expression were assessed after exposure to each hypoxic protocol. Additionally, cell viability was determined at baseline and after each glutamate challenge, in presence or absence of ceftriaxone that increases glutamate transporter expression. GFAP and MAP2 decreased after 7 days IH and SH. Long-term IH but not SH decreased EAAT1 and EAAT2. Excitotoxic glutamate challenge decreased cell viability and the following 200 µM exposure further increased cell death, particularly in IH-exposed slices. Ceftriaxone prevented glutamate transporter decrease and improved cell viability after IH and excitotoxicity. We conclude that IH is more detrimental to cell survival and glutamate homeostasis than SH. These findings suggest that impaired regulation of extracellular glutamate levels is implicated in the increased brain susceptibility to excitotoxic insult after long-term IH.


Subject(s)
Amino Acid Transport System X-AG/metabolism , Ceftriaxone/pharmacology , Cell Hypoxia/physiology , Cell Survival/physiology , Animals , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Glutamic Acid/pharmacology , In Vitro Techniques , Rats , Rats, Sprague-Dawley
4.
Free Radic Biol Med ; 47(10): 1440-9, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19703551

ABSTRACT

Heat shock protein 90 (Hsp90) is a chaperone protein regulating PC-12 cell survival by binding and stabilizing Akt, Raf-1, and Cdc37. Hsp90 inhibitor geldanamycin (GA) cytotoxicity has been attributed to the disruption of Hsp90 binding, and the contribution of oxidative stress generated by its quinone group has not been studied in this context. Reactive oxygen species (ROS) and cell survival were assessed in PC-12 cells exposed to GA or menadione (MEN), and Akt, Raf-1, and Cdc37 expression and binding to Hsp90 were determined. GA disrupted Hsp90 binding and increased ROS production starting at 1 h, and cell death occurred at 6 h, inhibited by N-acetylcysteine (NAC) without preventing dissociation of proteins. At 24 h, NAC prevented cytotoxicity and Hsp90 complex disruption. However, MnTBAP antioxidant treatment failed to inhibit GA cytotoxicity, suggesting that NAC acts by restoring glutathione. In contrast, 24 h MEN treatment induced cytotoxicity without disrupting Hsp90 binding. GA and MEN decreased Hsp90-binding protein expression, and proteasomal inhibition prevented MEN-, but not GA-induced degradation. In conclusion, whereas MEN cytotoxicity is mediated by ROS and proteasomal degradation, GA-induced cytotoxicity requires ROS but induces Hsp90 complex dissociation and proteasome-independent protein degradation. These differences between MEN- and GA-induced cytotoxicity may allow more specific targeting of cancer cells.


Subject(s)
Benzoquinones/toxicity , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/toxicity , Oxidative Stress/drug effects , Signal Transduction/drug effects , Animals , Cell Survival/drug effects , Drug Screening Assays, Antitumor , HSP90 Heat-Shock Proteins/metabolism , PC12 Cells , Proteasome Endopeptidase Complex/metabolism , Rats , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured , Vitamin K 3/pharmacology
5.
J Biol Chem ; 280(36): 31870-81, 2005 Sep 09.
Article in English | MEDLINE | ID: mdl-16027165

ABSTRACT

Hypoxia is a common environmental stress that influences signaling pathways and cell function. Previous studies from our laboratory have identified significant differences in cellular responses to sustained or intermittent hypoxia with the latter proving more cytotoxic. We hypothesized that differences in susceptibility of neurons to intermittent (IH) and sustained hypoxia (SH) are mediated by altered Akt signaling. SH, but not IH, induced a significant increase in Akt activation in rat CA1 hippocampal region extracts compared with room air controls. Akt immunoprecipitations followed by proteomic analysis identified valosin-containing protein (VCP) as an Akt-binding protein. In addition, VCP expression and association with Akt was enhanced during SH, and this association was decreased upon phosphoinositide 3-kinase/Akt pathway blockade with LY294002. Active recombinant Akt phosphorylated recombinant VCP in vitro. Site-directed mutagenesis studies identified Ser352, Ser746, and Ser748 as Akt phosphorylation sites on VCP. In addition, rat CA1 hippocampal tissue exposed to SH exhibited an acidic pI shift of VCP. Protein phosphatase 2A treatment inhibited this acidic shift consistent with SH-induced phosphorylation of VCP in vivo. PC-12 cells transfected with active Akt, but not dominant negative Akt or vector, induced VCP expression and an acidic shift in VCP pI, which was inhibited by protein phosphatase 2A treatment. Furthermore, VCP association with ubiquitinated proteins was demonstrated in vector-transfected PC-12 cell lysates, whereas active Akt-transfected cells demonstrated a marked decrease in association of VCP with ubiquitinated proteins. We concluded that Akt phosphorylates VCP in vitro and in vivo, and VCP phosphorylation releases it from ubiquitinated substrate protein(s) possibly allowing ubiquitinated protein(s) to be degraded by the proteosome.


Subject(s)
Cell Cycle Proteins/metabolism , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Ubiquitin/metabolism , Adenosine Triphosphatases , Animals , Brain/metabolism , Cell Cycle Proteins/chemistry , Hypoxia/metabolism , Isoelectric Point , Male , PC12 Cells , Phosphatidylinositol 3-Kinases/physiology , Proteasome Endopeptidase Complex/physiology , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , Serine/metabolism , Threonine/metabolism , Valosin Containing Protein
6.
J Appl Physiol (1985) ; 99(2): 642-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15817718

ABSTRACT

Tyrosine hydroxylase, a hypoxia-regulated gene, may be involved in tissue adaptation to hypoxia. Intermittent hypoxia, a characteristic feature of sleep apnea, leads to significant memory deficits, as well as to cortex and hippocampal apoptosis that are absent after sustained hypoxia. To examine the hypothesis that sustained and intermittent hypoxia induce different catecholaminergic responses, changes in tyrosine hydroxylase mRNA, protein expression, and activity were compared in various brain regions of male rats exposed for 6 h, 1 day, 3 days, and 7 days to sustained hypoxia (10% O(2)), intermittent hypoxia (alternating room air and 10% O(2)), or normoxia. Tyrosine hydroxylase activity, measured at 7 days, increased in the cortex as follows: sustained > intermittent > normoxia. Furthermore, activity decreased in the brain stem and was unchanged in other brain regions of sustained hypoxia-exposed rats, as well as in all regions from animals exposed to intermittent hypoxia, suggesting stimulus-specific and heterotopic catecholamine regulation. In the cortex, tyrosine hydroxylase mRNA expression was increased, whereas protein expression remained unchanged. In addition, significant differences in the time course of cortical Ser(40) tyrosine hydroxylase phosphorylation were present in the cortex, suggesting that intermittent and sustained hypoxia-induced enzymatic activity differences are related to different phosphorylation patterns. We conclude that long-term hypoxia induces site-specific changes in tyrosine hydroxylase activity and that intermittent hypoxia elicits reduced tyrosine hydroxylase recruitment and phosphorylation compared with sustained hypoxia. Such changes may not only account for differences in enzyme activity but also suggest that, with differential regional brain susceptibility to hypoxia, recruitment of different mechanisms in response to hypoxia will elicit region-specific modulation of catecholamine response.


Subject(s)
Brain/enzymology , Hypoxia/metabolism , Tyrosine 3-Monooxygenase/metabolism , Acute Disease , Adaptation, Physiological , Animals , Chronic Disease , Enzyme Activation , Gene Expression Regulation, Enzymologic , Hypoxia/classification , Male , Rats , Rats, Sprague-Dawley , Time Factors , Tissue Distribution
7.
Neurobiol Dis ; 17(1): 44-53, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15350964

ABSTRACT

Intermittent hypoxia (IH) during sleep induces significant neurobehavioral deficits in the rat. Since nitric oxide (NO) has been implicated in ischemia-reperfusion-related pathophysiological consequences, the temporal effects of IH (alternating 21% and 10% O(2) every 90 s) and sustained hypoxia (SH; 10% O(2)) during sleep for up to 14 days on the induction of nitric oxide synthase (NOS) isoforms in the brain were examined in the cortex of Sprague-Dawley rats. No significant changes of endothelial NOS (eNOS) and neuronal NOS (nNOS) occurred over time with either IH or SH. Similarly, inducible NOS (iNOS) was not affected by SH. However, increased expression and activity of iNOS were observed on days 1 and 3 of IH (P < 0.01 vs. control; n = 12/group) and were followed by a return to basal levels on days 7 and 14. Furthermore, IH-mediated neurobehavioral deficits in the water maze were significantly attenuated in iNOS knockout mice. We conclude that IH is associated with a time-dependent induction of iNOS and that the increased expression of iNOS may play a critical role in the early pathophysiological events leading to IH-mediated neurobehavioral deficits.


Subject(s)
Hypoxia, Brain/enzymology , Maze Learning/physiology , Memory Disorders/enzymology , Nitric Oxide Synthase/metabolism , Animals , Cerebral Cortex/chemistry , Cerebral Cortex/enzymology , Male , Mice , Mice, Knockout , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase/deficiency , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Rats , Rats, Sprague-Dawley
8.
Eur J Neurosci ; 18(8): 2335-42, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14622195

ABSTRACT

Intermittent hypoxia (IH) during sleep, such as occurs in obstructive sleep apnea, leads to degenerative changes in the hippocampus, and is associated with spatial learning deficits in the adult rat. We report that in Sprague-Dawley rats the initial IH-induced impairments in spatial learning are followed by a partial functional recovery over time, despite continuing IH exposure. These functional changes coincide with initial decreases in basal neurogenesis as shown by the number of positively colabelled cells for BrdU and neurofilament in the dentate gyrus of the hippocampus, and are followed by increased expression of neuronal progenitors and mature neurons (nestin and BrdU-neurofilament positively labelled cells, respectively). In contrast, no changes occurred during the course of IH exposures in the expression of the synaptic proteins synaptophysin, SNAP25, and drebrin. Collectively, these findings indicate that the occurrence of IH during the lights on period results in a biphasic pattern of neurogenesis in the hippocampus of adult rats, and may account for the observed partial recovery of spatial function.


Subject(s)
Hippocampus/pathology , Hypoxia/physiopathology , Neurons/metabolism , Recovery of Function , Spatial Behavior/physiology , Animals , Apoptosis , Blotting, Western , Bromodeoxyuridine/metabolism , Cues , Escape Reaction , Hippocampus/metabolism , Immunohistochemistry , Male , Maze Learning , Oxygen/metabolism , Rats , Rats, Sprague-Dawley , Reaction Time , Time Factors
9.
Hypertension ; 42(6): 1130-6, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14597643

ABSTRACT

Chronic intermittent hypoxia, a characteristic feature of sleep-disordered breathing, induces hypertension through augmented sympathetic nerve activity and requires the presence of functional carotid body arterial chemoreceptors. In contrast, chronic sustained hypoxia does not alter blood pressure. We therefore analyzed the biosynthetic pathways of catecholamines in peripheral nervous system structures involved in the pathogenesis of intermittent hypoxia-induced hypertension, namely, carotid bodies, superior cervical ganglia, and adrenal glands. Rats were exposed to either intermittent hypoxia (90 seconds of room air alternating with 90 seconds of 10% O2) or to sustained hypoxia (10% O2) for 1 to 30 days. Dopamine, norepinephrine, epinephrine, dihydroxyphenylacetic acid, and 5-hydroxytyptamine contents were measured by high-performance liquid chromatography. Expression of tyrosine hydroxylase and its phosphorylated forms, dopamine beta-hydroxylase, phenylethanolamine N-methyltransferase, and GTP cyclohydrolase-1 were determined by Western blot analyses. Both sustained and intermittent hypoxia significantly increased dopamine and norepinephrine content in carotid bodies but not in sympathetic ganglia or adrenal glands. In carotid bodies, both types of hypoxia augmented total levels of tyrosine hydroxylase protein and its phosphorylation on serines 19, 31, 40, as well as levels of GTP cyclohydrolase-1. However, the effects of intermittent hypoxia on catecholaminergic pathways were significantly smaller and delayed than those induced by sustained hypoxia. Thus, attenuated induction of catecholaminergic phenotype by intermittent hypoxia in carotid body may play a role in development of hypertension associated with sleep-disordered breathing. The effects of both types of hypoxia on expression of catecholaminergic enzymes in superior cervical neurons and adrenal glands were transient and small.


Subject(s)
Catecholamines/biosynthesis , Hypoxia/metabolism , Neurons/metabolism , Neurosecretory Systems/metabolism , Sympathetic Nervous System/metabolism , Adrenal Glands/metabolism , Animals , Blood Pressure , Carotid Body/metabolism , GTP Cyclohydrolase/metabolism , Hypertension/etiology , Hypoxia/complications , Hypoxia/physiopathology , Male , Neurosecretory Systems/cytology , Rats , Superior Cervical Ganglion/metabolism , Sympathetic Nervous System/cytology
10.
J Appl Physiol (1985) ; 95(5): 1767-74, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14555663

ABSTRACT

The effects of chronic sustained hypoxia (SH) on ventilation have been thoroughly studied. However, the effects of intermittent hypoxia (IH), a more prevalent condition in health and disease are currently unknown. We hypothesized that the ventilatory consequences of SH and IH may differ and be related to changes in N-methyl-D-aspartate (NMDA) glutamate receptor subunit expression. To examine these issues, Sprague-Dawley adult male rats were exposed to 30 days of either SH (10% O2) or IH (21% and 10% O2 alternations every 90 s) or to normoxia (RA), at the end of which ventilatory and O2 consumption responses to a 20-min acute hypoxic challenge (10% O2) were conducted. In addition, dorsocaudal brain stem tissue lysates were harvested at 1 h, 6 h, 1 day, 3 days, 7 days, 14 days, and 30 days of SH and IH and analyzed for NR1, NR2A, and NR2B NMDA glutamate receptor expression by immunoblotting. Normoxic ventilation was higher after both SH and IH (P < 0.001). Peak hypoxic ventilatory response was higher after SH but not after IH compared with RA. However, hypoxic ventilatory decline was more prominent after SH than IH (P < 0.001). NR1 expression showed a biphasic pattern of expression over time that was essentially identical after IH and SH (P value not significant). However, NR2A and NR2B expression was higher in IH compared with SH and RA (P < 0.01). We conclude that long-lasting exposures to SH and IH enhance normoxic ventilation but are associated with different time domains of ventilation during acute hypoxia that may be accounted in part by changes in NMDA glutamate receptor subunit expression.


Subject(s)
Brain Stem/metabolism , Hypoxia/metabolism , Hypoxia/physiopathology , Respiratory Mechanics/physiology , Animals , Body Weight , Chronic Disease , Male , Oxygen/pharmacology , Oxygen Consumption , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism
11.
FASEB J ; 17(12): 1709-11, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12958184

ABSTRACT

In neurons, hypoxia activates intracellular death-related pathways, yet the antiapoptotic mechanisms triggered by hypoxia remain unclear. In RN46A neuronal cells, minimum media growth conditions induced cell death as early as 12 h after the cells were placed in these conditions (i.e., after removal of B-27 supplement). However, apoptosis occurred in hypoxia (1% O2) only after 48 h, and in fact hypoxia reduced the apoptosis associated with trophic factor withdrawal. Furthermore, hypoxia induced time-dependent increases in expression of platelet-derived growth factor (PDGF) B mRNA and protein, as well as PDGF-beta receptor phosphorylation. Although exogenous PDGF-BB induced only transient Akt activation, hypoxia triggered persistent activation of Akt for up to 24 h. Inhibition of phosphatidylinositol 3-kinase (PI3K) or of PDGF-beta receptor phosphorylation abrogated both hypoxia-induced and exogenous PDGF-BB-induced Akt phosphorylation, and it completely abolished hypoxia-induced protection from media supplement deprivation, which suggests that the long-lasting activation of Akt during hypoxia and the prosurvival induction were due to endogenously generated PDGF-BB. Furthermore, these inhibitors decreased hypoxia-inducible factor 1alpha (HIF-1alpha) DNA binding, which suggests that the PDGF/PDGF-beta receptor/Akt pathway induces downstream HIF-1alpha gene transcription. We conclude that in RN46A neuronal cells, hypoxia activates an autocrine-paracrine antiapoptotic mechanism that involves up-regulation of PDGF-B and PDGF-beta receptor-dependent activation of the PI3K/Akt signaling pathway to induce downstream transcription of survival genes.


Subject(s)
Autocrine Communication , Neurons/metabolism , Paracrine Communication , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-sis/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Apoptosis , Cell Hypoxia , Cell Line , Cell Survival , Culture Media , Hypoxia-Inducible Factor 1, alpha Subunit , Kinetics , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Neurons/enzymology , Phosphorylation , Proto-Oncogene Proteins c-akt , Transcription Factors/biosynthesis
12.
Respir Physiol Neurobiol ; 136(2-3): 91-103, 2003 Jul 16.
Article in English | MEDLINE | ID: mdl-12853002

ABSTRACT

The CA1 and CA3 regions of the hippocampus markedly differ in their susceptibility to hypoxia in general, and more particularly to the intermittent hypoxia (IH) that characterizes sleep apnea. We used proteomic analysis to build a database of proteins expressed in normoxic CA1 and CA3. The current hippocampus protein database identifies 106 proteins. A hypothetical protein with accession number AK006737 (gimid R:12839969) was strongly upregulated in the CA1, but not CA3 hippocampal region. Bioinformatic analysis revealed that the unknown protein contained a high stringency protein kinase e binding site. Domain analysis demonstrated the presence of a conserved sequence indicative of macrophage scavenger receptors. Using proteomic analysis we have previously demonstrated that acute (6 h) IH-mediated CA1 injury results from complex interactions between pathways involving increased metabolism, induction of stress-induced proteins and apoptosis, and ultimately disruption of structural proteins and cell integrity. The current findings identify a hypothetical protein that may play a key role in the response of CA1 to IH. These findings provide initial insights into mechanisms underlying differences in susceptibility to hypoxia in neural tissue and demonstrate how proteomic analysis can be used to generate new hypotheses, which define neuronal adaptation to IH.


Subject(s)
Brain Chemistry , Hippocampus/metabolism , Hypoxia/metabolism , Nerve Tissue Proteins/isolation & purification , Proteomics/methods , Animals , Binding Sites , Blotting, Western , Computational Biology/methods , Databases, Protein , Electrophoresis, Gel, Two-Dimensional , Hippocampus/anatomy & histology , Male , Mass Spectrometry/methods , Nerve Tissue Proteins/metabolism , Protein Kinase C/metabolism , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Receptors, Immunologic/metabolism , Receptors, Scavenger , Sequence Analysis, Protein/methods , Time Factors
13.
Am J Respir Crit Care Med ; 168(4): 469-75, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12773326

ABSTRACT

Intermittent hypoxia (IH) during sleep, a critical feature of sleep apnea, induces significant neurobehavioral deficits in the rat. Cyclooxygenase (COX)-2 is induced during stressful conditions such as cerebral ischemia and could play an important role in IH-induced learning deficits. We therefore examined COX-1 and COX-2 genes and COX-2 protein expression and activity (prostaglandin E2 [PGE2] tissue concentration) in cortical regions of rat brain after exposure to either IH (10% O2 alternating with 21% O2 every 90 seconds) or sustained hypoxia (10% O2). In addition, the effect of selective COX-2 inhibition with NS-398 on IH-induced neurobehavioral deficits was assessed. IH was associated with increased COX-2 protein and gene expression from Day 1 to Day 14 of exposure. No changes were found in COX-1 gene expression after exposure to hypoxia. IH-induced COX-2 upregulation was associated with increased PGE2 tissue levels, neuronal apoptosis, and neurobehavioral deficits. Administration of NS-398 abolished IH-induced apoptosis and PGE2 increases without modifying COX-2 mRNA expression. Furthermore, NS-398 treatment attenuated IH-induced deficits in the acquisition and retention of a spatial task in the water maze. We conclude that IH induces upregulation and activation of COX-2 in rat cortex and that COX-2 may play a role in IH-mediated neurobehavioral deficits.


Subject(s)
Hypoxia/enzymology , Isoenzymes/analysis , Memory Disorders/enzymology , Peroxidases/analysis , Prostaglandin-Endoperoxide Synthases/analysis , Sleep Apnea Syndromes/enzymology , Analysis of Variance , Animals , Apoptosis/physiology , Cerebral Cortex/enzymology , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/analysis , Gene Expression Regulation, Enzymologic , Hypoxia/complications , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Male , Maze Learning , Membrane Proteins , Memory Disorders/etiology , Neurons/pathology , Nitrobenzenes/pharmacology , Peroxidases/genetics , Prostaglandin-Endoperoxide Synthases/genetics , Random Allocation , Rats , Rats, Sprague-Dawley , Sleep Apnea Syndromes/complications , Sulfonamides/pharmacology , Time Factors
14.
J Neurochem ; 83(2): 331-45, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12423243

ABSTRACT

The CA1 and CA3 regions of the hippocampus markedly differ in their susceptibility to hypoxia in general, and more particularly to the intermittent hypoxia that characterizes sleep apnea. Proteomic approaches were used to identify proteins differentially expressed in the CA1 and CA3 regions of the rat hippocampus and to assess changes in protein expression following a 6-h exposure to intermittent hypoxia (IH). Ninety-nine proteins were identified, and 15 were differentially expressed in the CA1 and the CA3 regions. Following IH, 32 proteins in the CA1 region and only 7 proteins in the more resistant CA3 area were up-regulated. Hypoxia-regulated proteins in the CA1 region included structural proteins, proteins related to apoptosis, primarily chaperone proteins, and proteins involved in cellular metabolic pathways. We conclude that IH-mediated CA1 injury results from complex interactions between pathways involving increased metabolism, induction of stress-induced proteins and apoptosis, and, ultimately, disruption of structural proteins and cell integrity. These findings provide initial insights into mechanisms underlying differences in susceptibility to hypoxia in neural tissue, and may allow for future delineation of interventional strategies aiming to enhance neuronal adaptation to IH.


Subject(s)
Hippocampus/chemistry , Hippocampus/metabolism , Hypoxia, Brain/metabolism , Proteome/analysis , Sleep Apnea, Obstructive/metabolism , Animals , Atmosphere Exposure Chambers , Blotting, Western , Disease Susceptibility/metabolism , Electrophoresis, Gel, Two-Dimensional , Male , Nerve Tissue Proteins/analysis , Rats , Rats, Sprague-Dawley
15.
J Biol Chem ; 277(38): 34708-16, 2002 Sep 20.
Article in English | MEDLINE | ID: mdl-12121987

ABSTRACT

Obstructive sleep apnea syndrome (OSAS), a disorder characterized by episodic hypoxia (EH) during sleep, is associated with systemic hypertension. We used proteomic analysis to examine differences in rat kidney protein expression during EH, and their potential relationship to EH-induced hypertension. Young male Sprague-Dawley rats were exposed to either EH or sustained hypoxia (SH) for 14 (EH14/SH14) and 30 (EH30/SH30) days. Mean arterial blood pressure was significantly increased only in EH30 (p < 0.0002). Kidney proteins were resolved by two-dimensional-PAGE and were identified by MALDI-MS. Renal expression of kallistatin, a potent vasodilator, was down-regulated in all animals. Expression of alpha-1-antitrypsin, an inhibitor of kallikrein activation, was up-regulated in EH but down-regulated in SH. Western blotting showed significant elevation of B(2)-bradykinin receptor expression in all normotensive animals but remained unchanged in hypertensive animals. Proteins relevant to vascular hypertrophy, such as smooth muscle myosin and protein-disulfide isomerase were up-regulated in EH30 but were down-regulated in SH30. These data indicate that EH induces changes in renal protein expression consistent with impairment of vasodilation mediated by the kallikrein-kallistatin pathway and vascular hypertrophy. In contrast, SH-induced changes suggest the kallikrein- and bradykinin-mediated compensatory mechanisms for prevention of hypertension and vascular remodeling. To test the hypothesis suggested by the proteomic data, we measured the effect of EH on blood pressure in transgenic hKLK1 rats that overexpress human kallikrein. Transgenic hKLK1 animals were protected from EH-induced hypertension. We conclude that EH-induced hypertension may result, at least in part, from altered regulation of the renal kallikrein system.


Subject(s)
Hypertension/metabolism , Hypoxia/complications , Kallikreins/metabolism , Kidney/metabolism , Proteome , Amino Acid Sequence , Animals , Blood Pressure , Electrophoresis, Gel, Two-Dimensional , Hypertension/etiology , Hypertension/physiopathology , Male , Molecular Sequence Data , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...