Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 10(5): e0124918, 2015.
Article in English | MEDLINE | ID: mdl-25933411

ABSTRACT

Cardiopulmonary arrest is a leading cause of death and disability in the United States that usually occurs in the aged population. Cardiac arrest (CA) induces global ischemia, disrupting global cerebral circulation that results in ischemic brain injury and leads to cognitive impairments in survivors. Ischemia-induced neuronal damage in the hippocampus following CA can result in the impairment of cognitive function including spatial memory. In the present study, we used a model of asphyxial CA (ACA) in nine month old male Fischer 344 rats to investigate cognitive and synaptic deficits following mild global cerebral ischemia. These experiments were performed with the goals of 1) establishing a model of CA in nine month old middle-aged rats; and 2) to test the hypothesis that learning and memory deficits develop following mild global cerebral ischemia in middle-aged rats. To test this hypothesis, spatial memory assays (Barnes circular platform maze and contextual fear conditioning) and field recordings (long-term potentiation and paired-pulse facilitation) were performed. We show that following ACA in nine month old middle-aged rats, there is significant impairment in spatial memory formation, paired-pulse facilitation n dysfunction, and a reduction in the number of non-compromised hippocampal Cornu Ammonis 1 and subiculum neurons. In conclusion, nine month old animals undergoing cardiac arrest have impaired survival, deficits in spatial memory formation, and synaptic dysfunction.


Subject(s)
Cognition Disorders/physiopathology , Heart Arrest/physiopathology , Hippocampus/physiopathology , Neuronal Plasticity , Aging , Animals , Asphyxia/complications , Asphyxia/physiopathology , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/physiopathology , Cognition Disorders/complications , Cognition Disorders/pathology , Conditioning, Psychological , Excitatory Postsynaptic Potentials , Fear , Freezing Reaction, Cataleptic , Heart Arrest/complications , Hippocampus/pathology , Male , Maze Learning , Memory Disorders/pathology , Memory Disorders/physiopathology , Rats, Inbred F344 , Survival Analysis , Synaptic Transmission
2.
J Cereb Blood Flow Metab ; 34(4): 613-20, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24447953

ABSTRACT

We previously showed that inhibition of protein kinase C delta (PKCδ) improves brain perfusion 24 hours after asphyxial cardiac arrest (ACA) and confers neuroprotection in the cortex and CA1 region of the hippocampus 7 days after arrest. Therefore, in this study, we investigate the mechanism of action of PKCδ-mediated hypoperfusion after ACA in the rat by using the two-photon laser scanning microscopy (TPLSM) to observe cortical cerebral blood flow (CBF) and laser Doppler flowmetry (LDF) detecting regional CBF in the presence/absence of δV1-1 (specific PKCδ inhibitor), nitric oxide synthase (NOS) substrate (L-arginine, L-arg) and inhibitor (N(ω)-Nitro-L-arginine, NLA), and nitric oxide (NO) donor (sodium nitroprusside, SNP). There was an increase in regional LDF and local (TPLSM) CBF in the presence of δV1-1+L-arg, but only an increase in regional CBF under δV1-1+SNP treatments. Systemic blood nitrite levels were measured 15 minutes and 24 hours after ACA. Nitrite levels were enhanced by pretreatment with δV1-1 30 minutes before ACA possibly attributable to enhanced endothelial NOS protein levels. Our results suggest that PKCδ can modulate NO machinery in cerebral vasculature. Protein kinase C delta can depress endothelial NOS blunting CBF resulting in hypoperfusion, but can be reversed with δV1-1 improving brain perfusion, thus providing subsequent neuroprotection after ACA.


Subject(s)
Blood Flow Velocity/physiology , Cerebral Cortex/blood supply , Cerebrovascular Circulation/physiology , Heart Arrest/enzymology , Nitric Oxide Synthase Type III/metabolism , Protein Kinase C-delta/antagonists & inhibitors , Animals , Arginine/pharmacology , Blood Flow Velocity/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebrovascular Circulation/drug effects , Enzyme Inhibitors/pharmacology , Heart Arrest/physiopathology , Laser-Doppler Flowmetry , Male , Microscopy, Confocal , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitrites/blood , Oligopeptides/pharmacology , Rats , Rats, Sprague-Dawley , Substrate Specificity
3.
Nitric Oxide ; 26(4): 241-50, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22484664

ABSTRACT

Cardiac arrest results in significant mortality after initial resuscitation due in most cases to ischemia-reperfusion induced brain injury and to a lesser degree myocardial dysfunction. Nitrite has previously been shown to protect against reperfusion injury in animal models of focal cerebral and heart ischemia. Nitrite therapy after murine cardiac arrest improved 22 h survival through improvements in myocardial contractility. These improvements accompanied transient mitochondrial inhibition which reduced oxidative injury to the heart. Based on preliminary evidence that nitrite may also protect against ischemic brain injury, we sought to test this hypothesis in a rat model of asphyxia cardiac arrest with prolonged survival (7d). Cardiac arrest resulted in hippocampal CA1 delayed neuronal death well characterized in this and other cardiac arrest models. Nitrite therapy did not alter post-arrest hemodynamics but did result in significant (75%) increases in CA1 neuron survival. This was associated with increases in hippocampal nitrite and S-nitrosothiol levels but not cGMP shortly after therapy. Mitochondrial function 1h after resuscitation trended towards improvement with nitrite therapy. Based on promising preclinical data, the first ever phase I trial of nitrite infusions in human cardiac arrest survivors has been undertaken. We present preliminary data showing low dose nitrite infusion did not result in hypotension or cause methemoglobinemia. Nitrite thus appears safe and effective for clinical translation as a promising therapy against cardiac arrest mediated heart and brain injury.


Subject(s)
Heart Arrest/physiopathology , Neuroprotective Agents/administration & dosage , Sodium Nitrite/administration & dosage , Animals , Blood Pressure/drug effects , Brain Ischemia/drug therapy , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/drug effects , Cyclic GMP/metabolism , Double-Blind Method , Heart Arrest/metabolism , Heart Arrest/pathology , Heart Rate/drug effects , Humans , Male , Methemoglobin/metabolism , Mitochondria/metabolism , Neuroprotective Agents/adverse effects , Random Allocation , Rats , Rats, Sprague-Dawley , S-Nitrosothiols/metabolism , Sodium Nitrite/adverse effects
4.
Circulation ; 120(10): 897-905, 2009 Sep 08.
Article in English | MEDLINE | ID: mdl-19704094

ABSTRACT

BACKGROUND: Three-fourths of cardiac arrest survivors die before hospital discharge or suffer significant neurological injury. Except for therapeutic hypothermia and revascularization, no novel therapies have been developed that improve survival or cardiac and neurological function after resuscitation. Nitrite (NO(2)(-)) increases cellular resilience to focal ischemia/reperfusion injury in multiple organs. We hypothesized that nitrite therapy may improve outcomes after the unique global ischemia/reperfusion insult of cardiopulmonary arrest. METHODS AND RESULTS: We developed a mouse model of cardiac arrest characterized by 12 minutes of normothermic asystole and a high cardiopulmonary resuscitation rate. In this model, global ischemia and cardiopulmonary resuscitation were associated with blood and organ nitrite depletion, reversible myocardial dysfunction, impaired alveolar gas exchange, neurological injury, and an approximately 50% mortality. A single low dose of intravenous nitrite (50 nmol=1.85 micromol/kg=0.13 mg/kg) compared with blinded saline placebo given at cardiopulmonary resuscitation initiation with epinephrine improved cardiac function, survival, and neurological outcomes. From a mechanistic standpoint, nitrite treatment restored intracardiac nitrite and increased S-nitrosothiol levels, decreased pathological cardiac mitochondrial oxygen consumption resulting from reactive oxygen species formation, and prevented oxidative enzymatic injury via reversible specific inhibition of respiratory chain complex I. CONCLUSIONS: Nitrite therapy after resuscitation from 12 minutes of asystole rapidly and reversibly modulated mitochondrial reactive oxygen species generation during early reperfusion, limiting acute cardiac dysfunction, death, and neurological impairment in survivors.


Subject(s)
Cardiopulmonary Resuscitation , Electron Transport Complex I/antagonists & inhibitors , Heart Arrest/physiopathology , Heart/physiopathology , Nervous System/physiopathology , Reactive Oxygen Species/antagonists & inhibitors , Sodium Nitrite/administration & dosage , Animals , Heart/drug effects , Heart Arrest/metabolism , Heart Arrest/therapy , Injections, Intravenous , Male , Mice , Mice, Inbred C57BL , Nervous System/drug effects , Sodium Nitrite/metabolism , Survival Rate , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...