Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Neuroscience ; 169(3): 1307-14, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20538047

ABSTRACT

Transient global brain ischemia results in an immediate inhibition of protein translation upon reperfusion. During early brain reperfusion protein synthesis is inhibited by alpha subunit of eukaryotic initiation factor 2 (eIF2alpha) phosphorylation by the PKR-like endoplasmic reticulum kinase (PERK). Normally, PERK is held in an inactive, monomeric state by the binding of the endoplasmic reticulum (ER) chaperone GRP78 to the lumenal end of PERK. The prevailing view is that ER stress leads to the accumulation of unfolded proteins in the ER lumen. GRP78 dissociates from PERK to bind these accumulated unfolded proteins, leading to PERK activation, phosphorylation of eIF2alpha, and inhibition of translation. To determine if an increase in unfolded nascent proteins following transient brain ischemia contributes to PERK activation, protein synthesis was blocked by intracerebral injection of anisomycin prior to induction of ischemia. Anisomycin inhibited protein synthesis by over 99% and reduced newly synthesized proteins in the ER to approximately 20% of controls. With an ER nearly devoid of newly synthesized proteins, PERK was still activated and was able to phosphorylate eIF2alpha in CA1 neurons during reperfusion. These data strongly argue that PERK activation is independent of the large increase in unfolded nascent proteins within the ER following transient global brain ischemia.


Subject(s)
Brain Ischemia/metabolism , Endoplasmic Reticulum/metabolism , Unfolded Protein Response , eIF-2 Kinase/metabolism , Animals , Anisomycin/pharmacology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , Endoplasmic Reticulum/drug effects , Enzyme Activation , Eukaryotic Initiation Factor-2/metabolism , Male , Molecular Chaperones/biosynthesis , Neurons/drug effects , Neurons/metabolism , Phosphorylation , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Long-Evans
2.
J Neurochem ; 77(5): 1418-21, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11389192

ABSTRACT

Reperfusion after global brain ischemia results initially in a widespread suppression of protein synthesis in neurons, which persists in vulnerable neurons, that is caused by the inhibition of translation initiation as a result of the phosphorylation of the alpha-subunit of eukaryotic initiation factor 2 (eIF2alpha). To identify kinases responsible for eIF2alpha phosphorylation [eIF2alpha(P)] during brain reperfusion, we induced ischemia by bilateral carotid artery occlusion followed by post-ischemic assessment of brain eIF2alpha(P) in mice with homozygous functional knockouts in the genes encoding the heme-regulated eIF2alpha kinase (HRI), or the amino acid-regulated eIF2alpha kinase (GCN2). A 10-fold increase in eIF2alpha(P) was observed in reperfused wild-type mice and in the HRI-/- or GCN2-/- mice. However, in all reperfused groups, the RNA-dependent protein kinase (PKR)-like endoplasmic reticulum eIF2alpha kinase (PERK) exhibited an isoform mobility shift on SDS-PAGE, consistent with the activation of the kinase. These data indicate that neither HRI nor GCN2 are required for the large increase in post-ischemic brain eIF2alpha(P), and in conjunction with our previous report that eIF2alpha(P) is produced in the brain of reperfused PKR-/- mice, provides evidence that PERK is the kinase responsible for eIF2alpha phosphorylation in the early post-ischemic brain.


Subject(s)
Brain Ischemia/metabolism , Reperfusion Injury/metabolism , eIF-2 Kinase/metabolism , Animals , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Fibroblasts/metabolism , Mice , Mice, Knockout , Phosphorylation , Precipitin Tests , eIF-2 Kinase/genetics
3.
J Neurol Sci ; 179(S 1-2): 1-33, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11054482

ABSTRACT

Brain ischemia and reperfusion engage multiple independently-fatal terminal pathways involving loss of membrane integrity in partitioning ions, progressive proteolysis, and inability to check these processes because of loss of general translation competence and reduced survival signal-transduction. Ischemia results in rapid loss of high-energy phosphate compounds and generalized depolarization, which induces release of glutamate and, in selectively vulnerable neurons (SVNs), opening of both voltage-dependent and glutamate-regulated calcium channels. This allows a large increase in cytosolic Ca(2+) associated with activation of mu-calpain, calcineurin, and phospholipases with consequent proteolysis of calpain substrates (including spectrin and eIF4G), activation of NOS and potentially of Bad, and accumulation of free arachidonic acid, which can induce depletion of Ca(2+) from the ER lumen. A kinase that shuts off translation initiation by phosphorylating the alpha-subunit of eukaryotic initiation factor-2 (eIF2alpha) is activated either by adenosine degradation products or depletion of ER lumenal Ca(2+). Early during reperfusion, oxidative metabolism of arachidonate causes a burst of excess oxygen radicals, iron is released from storage proteins by superoxide-mediated reduction, and NO is generated. These events result in peroxynitrite generation, inappropriate protein nitrosylation, and lipid peroxidation, which ultrastructurally appears to principally damage the plasmalemma of SVNs. The initial recovery of ATP supports very rapid eIF2alpha phosphorylation that in SVNs is prolonged and associated with a major reduction in protein synthesis. High catecholamine levels induced by the ischemic episode itself and/or drug administration down-regulate insulin secretion and induce inhibition of growth-factor receptor tyrosine kinase activity, effects associated with down-regulation of survival signal-transduction through the Ras pathway. Caspase activation occurs during the early hours of reperfusion following mitochondrial release of caspase 9 and cytochrome c. The SVNs find themselves with substantial membrane damage, calpain-mediated proteolytic degradation of eIF4G and cytoskeletal proteins, altered translation initiation mechanisms that substantially reduce total protein synthesis and impose major alterations in message selection, down-regulated survival signal-transduction, and caspase activation. This picture argues powerfully that, for therapy of brain ischemia and reperfusion, the concept of single drug intervention (which has characterized the approaches of basic research, the pharmaceutical industry, and clinical trials) cannot be effective. Although rigorous study of multi-drug protocols is very demanding, effective therapy is likely to require (1) peptide growth factors for early activation of survival-signaling pathways and recovery of translation competence, (2) inhibition of lipid peroxidation, (3) inhibition of calpain, and (4) caspase inhibition. Examination of such protocols will require not only characterization of functional and histopathologic outcome, but also study of biochemical markers of the injury processes to establish the role of each drug.


Subject(s)
Brain Ischemia/metabolism , Nerve Degeneration/metabolism , Reperfusion Injury/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis/physiology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Calpain/metabolism , Cell Differentiation/physiology , Cerebrovascular Circulation/physiology , Excitatory Amino Acids/metabolism , Free Radicals/metabolism , Genes, Immediate-Early/physiology , Growth Substances/metabolism , Humans , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Nerve Tissue Proteins/biosynthesis , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Signal Transduction/physiology
4.
J Cereb Blood Flow Metab ; 19(9): 1010-9, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10478653

ABSTRACT

Brain reperfusion causes prompt, severe, and prolonged protein synthesis suppression and increased phosphorylation of eukaryotic initiation factor 2alpha [eIF2alpha(P)] in hippocampal CA1 and hilar neurons. The authors hypothesized that eIF2alpha(P) dephosphorylation would lead to recovery of protein synthesis. Here the effects of insulin, which activates phosphatases, were examined by immunostaining for eIF2alpha(P) and autoradiography of in vivo 35S amino acid incorporation. Rats resuscitated from a 10-minute cardiac arrest were given 0, 2, 10 or 20 U/kg of intravenous insulin, underwent reperfusion for 90 minutes, and were perfusion fixed. Thirty minutes before perfusion fixation, control and resuscitated animals received 500 microCi/kg of 35S methionine/cysteine. Alternate 30-microm brain sections were autoradiographed or immunostained for eIF2alpha(P). Controls had abundant protein synthesis and no eIF2alpha(P) in hippocampal neurons. Untreated reperfused neurons in the CA1, hilus, and dentate gyrus had intense staining for eIF2alpha(P) and reduced protein synthesis; there was little improvement with treatment with 2 or 10 U/kg of insulin. However, with 20 U/kg of insulin, these neurons recovered protein synthesis and were free of eIF2alpha(P). These results show that the suppression of protein synthesis in the reperfused brain is reversible; they support a causal association between eIF2alpha(P) and inhibition of protein synthesis, and suggest a mechanism for the neuroprotective effects of insulin.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Hippocampus/metabolism , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Ischemic Attack, Transient/metabolism , Nerve Tissue Proteins/biosynthesis , Animals , Autoradiography , Hippocampus/blood supply , Hippocampus/pathology , Ischemic Attack, Transient/pathology , Male , Neurons/metabolism , Neurons/pathology , Phosphorylation , Rats , Rats, Long-Evans
5.
Exp Neurol ; 155(2): 221-7, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10072297

ABSTRACT

When ischemic brain is reperfused, there is in vulnerable neurons immediate inhibition of protein synthesis associated with a large increase in phosphorylation of the alpha-subunit of eukaryotic initiation factor 2 [eIF2alpha, phosphorylated form eIF2alpha(P)]. We examined eIF2alpha kinase and eIF2alpha(P) phosphatase activity in brain homogenate postmitochondrial supernatants obtained from rats after 3 to 30 min of global brain ischemia (cardiac arrest), after 5 min of ischemia and 5 min of reperfusion (5R), and after 10 min of ischemia and 90 min reperfusion (90R). Because it has been suggested that PKR might be specifically responsible for producing eIF2alpha(P) during reperfusion, we also examined in brain homogenates from wild-type and PKR0/0 C57BL/6J x 129/SV mice the effect of 5 min of ischemia and 5 min of reperfusion on eIF2alpha(P). Cytosolic brain eIF2alpha(P) in the 5R and 90R rats was 18- and 23-fold that of nonischemic controls without any change in the rate of eIF2alpha(P) dephosphorylation. There was no change in eIF2alpha kinase activity between 3 and 30 min of ischemia but an 85% decrease in the 5R group; the 90R group was similar to controls. In wild-type and PKR0/0 mice total eIF2alpha was identical, and there was an identical 16-fold increase in eIF2alpha(P) at 5 min of reperfusion. Our observations contradict hypotheses that PKR activation, loss of eIF2alpha(P) phosphatase activity, or any general increase in eIF2alpha kinase activity are responsible for reperfusion-induced phosphorylation of eIF2alpha, and we suggest that the mechanism may involve regulation of the availability of eIF2alpha to a kinase.


Subject(s)
Ischemic Attack, Transient/enzymology , Phosphoprotein Phosphatases/metabolism , Reperfusion Injury/enzymology , eIF-2 Kinase/metabolism , Animals , Autoradiography , Blotting, Western , Brain/enzymology , Mice , Mice, Inbred C57BL , Phosphoprotein Phosphatases/biosynthesis , Phosphorylation , Rats , Rats, Long-Evans , eIF-2 Kinase/biosynthesis
6.
Proc Natl Acad Sci U S A ; 95(24): 14511-6, 1998 Nov 24.
Article in English | MEDLINE | ID: mdl-9826731

ABSTRACT

Protein synthesis (PS) has been considered essential to sustain mammalian life, yet was found to be virtually arrested for weeks in brain and other organs of the hibernating ground squirrel, Spermophilus tridecemlineatus. PS, in vivo, was below the limit of autoradiographic detection in brain sections and, in brain extracts, was determined to be 0.04% of the average rate from active squirrels. Further, it was reduced 3-fold in cell-free extracts from hibernating brain at 37 degreesC, eliminating hypothermia as the only cause for protein synthesis inhibition (active, 0.47 +/- 0.08 pmol/mg protein per min; hibernator, 0.16 +/- 0.05 pmol/mg protein per min, P < 0.001). PS suppression involved blocks of initiation and elongation, and its onset coincided with the early transition phase into hibernation. An increased monosome peak with moderate ribosomal disaggregation in polysome profiles and the greatly increased phosphorylation of eIF2alpha are both consistent with an initiation block in hibernators. The elongation block was demonstrated by a 3-fold increase in ribosomal mean transit times in cell-free extracts from hibernators (active, 2.4 +/- 0.7 min; hibernator, 7.1 +/- 1.4 min, P < 0.001). No abnormalities of ribosomal function or mRNA levels were detected. These findings implicate suppression of PS as a component of the regulated shutdown of cellular function that permits hibernating ground squirrels to tolerate "trickle" blood flow and reduced substrate and oxygen availability. Further study of the factors that control these phenomena may lead to identification of the molecular mechanisms that regulate this state.


Subject(s)
Brain/metabolism , Hibernation/physiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Peptide Chain Elongation, Translational , Peptide Chain Initiation, Translational , Protein Biosynthesis , Ribosomes/metabolism , Adenosine Diphosphate Ribose/metabolism , Animals , Autoradiography/methods , Carbon Radioisotopes , Eukaryotic Initiation Factor-2/metabolism , Leucine/metabolism , Nerve Tissue Proteins/isolation & purification , Peptide Elongation Factor 2 , Peptide Elongation Factors/metabolism , Phosphorylation , RNA, Messenger/metabolism , Sciuridae , Sensitivity and Specificity , Transcription, Genetic
7.
J Cereb Blood Flow Metab ; 18(8): 876-81, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9701348

ABSTRACT

Global brain ischemia and reperfusion result in the degradation of the eukaryotic initiation factor (eIF) 4G, which plays a critical role in the attachment of the mRNA to the ribosome. Because eIF-4G is a substrate of calpain, these studies were undertaken to examine whether calpain I activation during global brain ischemia contributes to the degradation of eIF-4G in vivo. Immunoblots with antibodies against calpain I and eIF-4G were prepared from rat brain postmitochondrial supernatant incubated at 37 degrees C with and without the addition of calcium and the calpain inhibitors calpastatin or MDL-28,170. Addition of calcium alone resulted in calpain I activation (as measured by autolysis of the 80-kDa subunit) and degradation of eIF-4G; this effect was blocked by either 1 micromol/L calpastatin or 10 micromol/L MDL-28,170. In rabbits subjected to 20 minutes of cardiac arrest, immunoblots of brain postmitochondrial supernatants showed that the percentage of autolyzed calpain I increased from 1.9% +/- 1.1% to 15.8% +/- 5.0% and that this was accompanied by a 68% loss of eIF-4G. MDL-28,170 pretreatment (30 mg/kg) decreased ischemia-induced calpain I autolysis 40% and almost completely blocked eIF-4G degradation. We conclude that calpain I degrades eIF-4G during global brain ischemia.


Subject(s)
Brain/metabolism , Calpain/metabolism , Ischemic Attack, Transient/metabolism , Peptide Initiation Factors/metabolism , Animals , Brain/drug effects , Calcium/pharmacology , Calcium-Binding Proteins/pharmacology , Calpain/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Eukaryotic Initiation Factor-4G , Female , Kinetics , Male , Rabbits , Rats , Reperfusion , Subcellular Fractions/metabolism
8.
J Cereb Blood Flow Metab ; 17(12): 1291-302, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9397028

ABSTRACT

Postischemic brain reperfusion is associated with a substantial and long-lasting reduction of protein synthesis in selectively vulnerable neurons. Because the overall translation initiation rate is typically regulated by altering the phosphorylation of serine 51 on the alpha-subunit of eukaryotic initiation factor 2 (eIF-2 alpha), we used an antibody specific to phosphorylated eIF-2 alpha [eIF-2(alpha P)] to study the regional and cellular distribution of eIF-2(alpha P) in normal, ischemic, and reperfused rat brains. Western blots of brain postmitochondrial supernatants revealed that approximately 1% of all eIF-2 alpha is phosphorylated in controls, eIF-2(alpha P) is not reduced by up to 30 minutes of ischemia, and eIF-2(alpha P) is increased approximately 20-fold after 10 and 90 minutes of reperfusion. Immunohistochemistry shows localization of eIF-2(alpha P) to astrocytes in normal brains, a massive increase in eIF-2(alpha P) in the cytoplasm of neurons within the first 10 minutes of reperfusion, accumulation of eIF-2(alpha P) in the nuclei of selectively vulnerable neurons after 1 hour of reperfusion, and morphology suggesting pyknosis or apoptosis in neuronal nuclei that continue to display eIF-2(alpha P) after 4 hours of reperfusion. These observations, together with the fact that eIF-2(alpha P) inhibits translation initiation, make a compelling case that eIF-2(alpha P) is responsible for reperfusion-induced inhibition of protein synthesis in vulnerable neurons.


Subject(s)
Brain Ischemia/metabolism , Reperfusion Injury/metabolism , eIF-2 Kinase/metabolism , Animals , Immunohistochemistry , Male , Phosphorylation , Rats , eIF-2 Kinase/analysis
9.
J Neurochem ; 67(5): 2005-12, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8863507

ABSTRACT

We used in vitro translation and antibodies against phosphoserine and the eukaryotic initiation factors elF-4E, elF-4G, and elF-2 alpha to examine the effects of global brain ischemia and reperfusion on translation initiation and its regulation in a rat model of 10 min of cardiac arrest followed by resuscitation and 90 min of reperfusion. Translation reactions were performed on postmitochondrial supernatants from brain homogenates with and without aurintricarboxylic acid to separate incorporation due to run-off from incorporation due to peptide synthesis initiated in vitro. The rate of leucine incorporation due to in vitro-initiated protein synthesis in normal forebrain homogenates was approximately 0.4 fmol of leucine/min/microgram of protein and was unaffected by 10 min of cardiac arrest, but 90 min of reperfusion reduced this rate 83%. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis and western blots of these homogenates showed that neither 10 min of global brain ischemia nor 90 min of reperfusion induced significant alterations in the quantity or serine phosphorylation of elF-4E. However, we observed in all 90-min-reperfused samples elF-4G fragments that also bound elF-4E. The amount of elF-2 alpha was not altered by ischemia or reperfusion, and immunoblotting after isoelectric focusing did not detect serine-phosphorylated elF-2 alpha in normal samples or in those obtained after ischemia without reperfusion. However, serine-phosphorylated elF-2 alpha was uniformly present after 90 min of reperfusion and represented 24 +/- 3% of the elF-2 alpha in these samples. The serine phosphorylation of elF-2 alpha and partial fragmentation of elF-4G observed after 90 min of reperfusion offer an explanation for the inhibition of protein synthesis.


Subject(s)
Eukaryotic Initiation Factor-2/biosynthesis , Ischemic Attack, Transient/metabolism , Peptide Chain Initiation, Translational , Peptide Initiation Factors/biosynthesis , Protein Biosynthesis , Animals , Antibodies , Blotting, Western , Eukaryotic Initiation Factor-4E , Gene Expression Regulation , Heart Arrest , Kinetics , Male , Phosphorylation , Phosphoserine/analysis , Rats , Reperfusion , Resuscitation , Subcellular Fractions/metabolism
10.
Ann Emerg Med ; 27(5): 588-94, 1996 May.
Article in English | MEDLINE | ID: mdl-8629779

ABSTRACT

Brain damage accompanying cardiac arrest and resuscitation is frequent and devastating. Neurons in the hippocampus CA1 and CA4 zones and cortical layers III and V are selectively vulnerable to death after injury by ischemia and reperfusion. Ultrastructural evidence indicates that most of the structural damage is associated with reperfusion, during which the vulnerable neurons develop disaggregation of polyribosomes, peroxidative damage to unsaturated fatty acids in the plasma membrane, and prominent alterations in the structure of the Golgi apparatus that is responsible for membrane assembly. Reperfusion is also associated with vulnerable neurons with prominent production of messenger RNAs for stress proteins and for the proteins of the activator protein-1 complex, but these vulnerable neurons fail to efficiently translate these messages into the proteins. The inhibition of protein synthesis during reperfusion involves alteration of translation initiation factors, specifically serine phosphorylation of the alpha-subunit of eukaryotic initiation factor-2 (elF-2 alpha). Growth factors--in particular, insulin--have the potential to reverse phosphorylation of elF-2 alpha, promote effective translation of the mRNA transcripts generated in response to ischemia and reperfusion, enhance neuronal defenses against radicals, and stimulate lipid synthesis and membrane repair. There is now substantial evidence that the insulin-class growth factors have neuron-sparing effects against damage by radicals and ischemia and reperfusion. This new knowledge may provide a fundamental basis for a rational approach to "cerebral resuscitation" that will allow substantial amelioration of the often dismal neurologic outcome now associated with resuscitation from cardiac arrest.


Subject(s)
Brain Ischemia/etiology , Cardiopulmonary Resuscitation , Heart Arrest/complications , Reperfusion Injury/etiology , Brain Ischemia/metabolism , Brain Ischemia/therapy , Growth Substances/therapeutic use , Hippocampus/blood supply , Hippocampus/injuries , Humans , Oxidative Stress/physiology , Protein Biosynthesis , Reperfusion Injury/metabolism , Reperfusion Injury/therapy , Risk Factors
11.
J Neurochem ; 66(1): 421-4, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8522983

ABSTRACT

Proteolytic degradation of numerous calpain substrates, including cytoskeletal and regulatory proteins, has been observed during brain ischemia and reperfusion. In addition, calpain inhibitors have been shown to decrease degradation of these proteins and decrease postischemic neuronal death. Although these observations support the inference of a role for mu-calpain in the pathophysiology of ischemic neuronal injury, the evidence is indirect. A direct indicator of mu-calpain proteolytic activity is autolysis of its 80-kDa catalytic subunit, and therefore we examined the mu-calpain catalytic subunit for evidence of autolysis during cerebral ischemia. Rabbit brain homogenates obtained after 0, 5, 10, and 20 min of cardiac arrest were electrophoresed and immunoblotted with a monoclonal antibody specific to the mu-calpain catalytic subunit. In nonischemic brain homogenates the antibody identified an 80-kDa band, which migrated identically with purified mu-calpain, and faint 78- and 76-kDa bands, which represent autolyzed forms of the 80-kDa subunit. The average density of the 80-kDa band decreased by 25 +/- 4 (p = 0.008) and 28 +/- 9% (p = 0.004) after 10 and 20 min of cardiac arrest, respectively, whereas the average density of the 78-kDa band increased by 111 +/- 50% (p = 0.02) after 20 min of cardiac arrest. No significant change in the density of the 76-kDa band was detected. These results provide direct evidence for autolysis of brain mu-calpain during cerebral ischemia. Further work is needed to characterize the extent, duration, and localization of mu-calpain activity during brain ischemia and reperfusion as well as its role in the causal pathway of postischemic neuronal injury.


Subject(s)
Brain Ischemia/metabolism , Calpain/metabolism , Isoenzymes/metabolism , Nerve Tissue Proteins/metabolism , Animals , Autolysis , Blotting, Western , Female , Rabbits
12.
J Neurochem ; 65(3): 1391-4, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7643117

ABSTRACT

Suppression of protein synthesis in the brain following an ischemic insult has been thought to occur because of inhibition of translation initiation. All eukaryotic mRNAs, with the exception of heat-shock transcripts, require the activity of eukaryotic initiation factor (eIF) 4E for formation of the translation initiation complex, and eIF-4E availability is rate-limiting. The response of brain eIF-4E concentration and phosphorylation following decapitation ischemia was studied in rat brain homogenates after electrophoresis and western blotting with antibodies against eIF-4E and phosphoserine, respectively. There was no change in level of eIF-4E after 5 min of ischemia (p = 0.82 vs. time 0), but it had decreased 32 (p = 0.01) and 57% (p = 0.006) after 10 and 20 min of ischemia, respectively. There was no loss of serine phosphorylation on eIF-4E beyond signal loss observed due to degradation of the protein itself (p = 0.31). In vitro exposure of eIF-4E to activated mu-calpain resulted in a 50% loss in 10 min of eIF-4E on western blots. If active eIF-4E is required for translation of its own mRNA, degradation of this protein during ischemia, possibly by activated mu-calpain, could be a direct mechanism of irreversible neuronal injury, and the rate of proteolysis of eIF-4E could place an upper time limit on the maximal duration of global brain ischemia compatible with neurologic recovery.


Subject(s)
Ischemic Attack, Transient/metabolism , Peptide Initiation Factors/metabolism , Animals , Blotting, Western , Brain/metabolism , Calpain/pharmacology , Eukaryotic Initiation Factor-4E , Male , Peptide Initiation Factors/genetics , Phosphorylation , Phosphoserine/metabolism , RNA, Messenger/metabolism , Rats , Time Factors
13.
Ann Emerg Med ; 25(6): 809-17, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7755206

ABSTRACT

STUDY HYPOTHESIS: We attempted to determine whether the reduced egress of mRNA from brain nuclei following in vivo ischemia and reperfusion is caused by direct damage to the nuclear pore-associated NTPase that impairs the system for nuclear export of polyadenylated, or poly(A)+, mRNA. DESIGN: Prospective animal study. INTERVENTIONS: NTPase activity and poly(A)+ mRNA transport were studied in nuclear envelope vesicles (NEVs) prepared from canine parietal cortex isolated after 20 minutes of ischemia or 20 minutes of ischemia and 2 or 6 hours of reperfusion. RESULTS: Brain NEV NTPase Michaelis-Menten constant (Km) and maximum uptake velocity (Vmax) and the ATP-stimulated poly(A)+ mRNA egress rates were not significantly affected by ischemia and reperfusion. In vitro exposure of the NEVs to the OH. radical-generating system completely abolished NTPase activity. CONCLUSION: We conclude that brain ischemia and reperfusion do not induce direct inhibition of nucleocytoplasmic transport of poly(A)+ mRNA. This suggests that the nuclear membrane is not exposed to significant concentrations of OH. radical during reperfusion.


Subject(s)
Acid Anhydride Hydrolases/pharmacokinetics , Brain Ischemia/metabolism , RNA, Messenger/metabolism , Reperfusion , Animals , Biological Transport , Dogs , Heart Arrest/metabolism , Hydroxyl Radical/metabolism , Nuclear Envelope/metabolism , Nucleoside-Triphosphatase , Prospective Studies
14.
Acta Neuropathol ; 90(1): 17-30, 1995.
Article in English | MEDLINE | ID: mdl-7572075

ABSTRACT

The neocortex and the hippocampus were examined for lipid peroxidation products and ultrastructural alterations by fluorescence and electron microscopy, respectively, in rats subjected to 10 min of cardiac arrest or 10 min cardiac arrest and either 90 or 360 min reperfusion. Lipid peroxidation products were observed after 90 min reperfusion in the perikarya and proximal dendrites of neocortical pyramidal neurons and in the hippocampal hilar cells and CA1, region; the fluorescence was most intense at the base of the apical dendrite, the region of the Golgi apparatus. After 90 min of reperfusion, the CA1, showed considerable stretches of rough endoplasmic reticulum devoid of ribosomes and the Golgi cisternae were shorter and widely dilated. The neocortex showed similar endoplasmic reticulum changes, but no significant alterations to the Golgi were noted. In addition there were areas where strings of ribosomes appear to be detaching from the endoplasmic reticulum. After 360 min reperfusion in both the neocortex and the hippocampus, the damage appeared more severe. The Golgi was fragmented into vacuoles, membranous whorls had appeared, and dense aggregates of smooth vesicles were seen coalescing with each other and the vacuoles. These observations suggest that early Golgi involvement is a more important marker of lethal injury than ribosome release from the endoplasmic reticulum. The areas of disturbed Golgi ultrastructure correspond to those areas that show evidence of lipid peroxidation and imply that lipid peroxidation may be causally related to the disturbance in Golgi ultrastructure.


Subject(s)
Brain Ischemia/metabolism , Golgi Apparatus/ultrastructure , Hippocampus/ultrastructure , Neurons/ultrastructure , Animals , Cell Death , Fluorescence , Heart Arrest , Lipid Peroxidation , Microscopy, Electron , Rats , Rats, Inbred Strains , Reperfusion
15.
Exp Neurol ; 124(2): 351-6, 1993 Dec.
Article in English | MEDLINE | ID: mdl-7507063

ABSTRACT

Rat brain nuclear proteins were examined for tyrosine phosphorylation after resuscitation from a 10-min cardiac arrest. Insulin (1 unit/kg intravenously), given immediately after resuscitation, caused a marked increase in tyrosine phosphorylation of a 90-kDa brain protein. This effect occurred without hypoglycemia and was not observed after insulin administration in previously insulinopenic, diabetic, nonischemic animals. Insulin-responsive tyrosine phosphorylation of a specific 90-kDa protein during reperfusion may represent insulin stimulation of a neuroprotective brain response to an ischemic insult, consistent with recent observations that insulin administration during reperfusion protects selectively vulnerable neurons from postischemic death.


Subject(s)
Brain/metabolism , Heart Arrest/metabolism , Insulin/pharmacology , Ischemic Attack, Transient/metabolism , Nerve Tissue Proteins/metabolism , Reperfusion , Analysis of Variance , Animals , Brain/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Diabetes Mellitus, Experimental/metabolism , Heart Arrest/physiopathology , Male , Myocardial Reperfusion , Phosphoproteins/isolation & purification , Phosphoproteins/metabolism , Phosphorylation , Phosphotyrosine , Rats , Rats, Wistar , Resuscitation , Tyrosine/analogs & derivatives , Tyrosine/analysis , Tyrosine/metabolism
18.
Ann Emerg Med ; 22(6): 970-9, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8503535

ABSTRACT

Rotational acceleration from closed-head trauma produces shear-strain brain injury at the interface of gray and white matter. The initial injury is followed by progressive damage involving three key phenomena: progression of subtle focal axonal damage to axonal transection between six and 12 hours after injury, progressive development of tissue microhemorrhages between 12 and 96 hours after injury, and development of tissue and cerebral spinal fluid lactic acidosis that does not appear to be explained by trauma-induced tissue depolarization, activation of phospholipases and the release of free arachidonic acid, radical generation by metabolism of arachidonate, and lipid peroxidation with consequent membrane degradation and partial mitochondrial uncoupling. Because of terminal differentiation, neurons may have a limited membrane repair capability that might be stimulated by growth factors. Other potential therapeutic interventions include calmodulin inhibitors, iron chelators, and free radical scavengers.


Subject(s)
Brain Injuries/drug therapy , Brain Injuries/physiopathology , Animals , Humans
19.
Stroke ; 24(5): 747-55; discussion 755-6, 1993 May.
Article in English | MEDLINE | ID: mdl-8488532

ABSTRACT

BACKGROUND: Brain ischemia and reperfusion produce profound protein synthesis alterations, the extent and persistence of which are dependent on the nature of the ischemia, the brain region, the cell layer within a region, and the particular proteins studied. After transient ischemia, most brain regions recover their protein synthesis capability; however, recovery in the selectively vulnerable areas is poor. It is unknown whether this phenomenon itself provokes or is a consequence of the process of neuronal death. SUMMARY OF REVIEW: Protein synthesis suppression during ischemia is due to energy depletion, but this is quickly reversed upon recirculation. Reperfusion does not appear to damage DNA or transcription mechanisms, although there are changes in the profile of transcripts being made. Similarly, purified ribosomes isolated from reperfused brains can make the normal repertoire of proteins and heat-shock proteins. However, during early reperfusion, newly synthesized messenger RNAs appear to accumulate in the nucleus; this alteration in RNA handling could reflect disruption at any of several steps, including posttranscriptional processing, nuclear pore transport, cytoskeletal binding, or formation of the translation initiation complex. Another mechanism that may be responsible for protein synthesis suppression during late reperfusion is progressive membrane destruction, with consequent shifts in the concentration of ions crucial for ribosomal function. CONCLUSIONS: Protein synthesis suppression after ischemia likely involves a progression of multiple mechanisms during reperfusion. Although the recent work reviewed here offers new insight into the potential mechanisms disrupting protein synthesis, detailed understanding will require further investigation.


Subject(s)
Brain Chemistry , Brain Ischemia/metabolism , Protein Biosynthesis , Animals , Brain Ischemia/pathology , Cell Death , Neurons , Proto-Oncogene Proteins/biosynthesis , RNA, Messenger/metabolism , Reperfusion
20.
Resuscitation ; 25(2): 161-70, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8493403

ABSTRACT

Previous studies have demonstrated that brain protein synthesis declines after global ischemia and reperfusion. To investigate the role of the translation system in this phenomenon, we examined the ability of partially purified ribosomes, ribosome-bound mRNA and translation cofactors derived from the transiently ischemic cerebral cortex to synthesize protein in vitro. Samples were prepared from canines subjected to 20-min cardiac arrest and after 2 or 8 h of post-resuscitation intensive care. There was no significant decrease in the rate of in vitro protein synthesis as a consequence of either ischemia or reperfusion. Northern hybridization of ribosome-bound RNA revealed a discrete band of mRNA for brain-specific creatine kinase (ck-bb) that was consistent in presence and intensity in all groups. However, mRNA for heat shock 70 protein (hsp-70) was observed only during reperfusion and markedly increased between 2 and 8 h reperfusion. Thus, we conclude that (1) the transcription system is intact during reperfusion and hsp-70 mRNA is made and translocated to the ribosomes during reperfusion, (2) mRNA for ck-bb is not displaced from ribosomes by the appearance of hsp-70 during reperfusion and (3) isolated ribosomes maintain their ability to translate in vitro during the first 8 h of reperfusion after global brain ischemia. Therefore, the early reduction in protein synthesis observed in vivo during post-ischemic brain reperfusion is not due to an intrinsic dysfunction of the ribosomes.


Subject(s)
Brain Ischemia/metabolism , Cerebral Cortex/metabolism , Creatine Kinase/biosynthesis , Heat-Shock Proteins/biosynthesis , RNA, Messenger/biosynthesis , Reperfusion Injury/metabolism , Animals , Blotting, Northern , Brain Ischemia/genetics , Creatine Kinase/genetics , Dogs , Heart Arrest/therapy , Heat-Shock Proteins/genetics , Isoenzymes , Protein Biosynthesis/physiology , Reperfusion Injury/genetics , Resuscitation , Ribosomes/metabolism , Transcription, Genetic/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...