Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Rev Esp Cardiol (Engl Ed) ; 73(2): 123-130, 2020 Feb.
Article in English, Spanish | MEDLINE | ID: mdl-30857978

ABSTRACT

INTRODUCTION AND OBJECTIVES: To analyze neuron-specific enolase (NSE) kinetics as a prognostic biomarker of neurological outcome in cardiac arrest survivors treated with targeted temperature management. METHODS: We performed a retrospective analysis of patients resuscitated from in- or out-of-hospital cardiac arrest admitted from September 2006 to May 2018 in a single tertiary care center and cooled to 32°C to 34°C for 24 hours. Blood samples for measurement of NSE values were drawn at hospital admission and at 24, 48, and 72hours after return of spontaneous circulation (ROSC). Neurological outcome was evaluated by means of the Cerebral Performance Category (CPC) score at 3 months and was characterized as good (CPC 1-2) or poor (CPC 3-5). RESULTS: Of 451 patients, 320 fulfilled the inclusion criteria and were analyzed (80.3% male, mean age 61±14.1 years). Among these, 174 patients (54.4%) survived with good neurological status. Poor outcome patients had higher median NSE values at hospital admission and at 24, 48 and 72 hours after ROSC. At 48 and 72 hours after ROSC, NSE predicted poor neurological outcome with areas under the receiver-operating characteristic curves of 0.85 (95%CI, 0.81-0.90) and 0.88 (95%CI, 0.83-0.93), respectively. In addition, delta NSE values between 72hours after ROSC and hospital admission predicted poor neurological outcome with an area under the receiver-operating characteristic curve of 0.90 (95%CI, 0.85-0.95) and was an independent predictor of unfavorable outcome on multivariate analysis (P <.001). CONCLUSIONS: In cardiac arrest survivors treated with targeted temperature management, delta NSE values between 72 hours after ROSC and hospital admission strongly predicted poor neurological outcome.


Subject(s)
Heart Arrest/enzymology , Nervous System Diseases/enzymology , Phosphopyruvate Hydratase/blood , Postoperative Complications/epidemiology , Biomarkers/blood , Female , Follow-Up Studies , Heart Arrest/complications , Heart Arrest/mortality , Humans , Incidence , Male , Middle Aged , Nervous System Diseases/epidemiology , Nervous System Diseases/etiology , Prognosis , ROC Curve , Retrospective Studies , Spain/epidemiology , Survival Rate/trends
2.
Int J Mol Med ; 43(4): 1575-1584, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30816437

ABSTRACT

Acute pulmonary embolism (APE) with cardiac arrest (CA) is associated with a high mortality rate. Even upon return of the spontaneous circulation (ROSC), APE­CA survivors are prone to myocardial cell apoptosis, a key cellular mechanism that induces heart failure. A recent study by our group discovered a post­resuscitation imbalance in the serum angiotensin­converting enzyme (ACE)2/ACE axis of the renin­angiotensin system (RAS), as well as regressive cardiac function in a porcine model of APE­CA. However, it has remained elusive how this imbalance in the ACE2/ACE axis affects myocardial cell apoptosis. In the present study, western blot and immunohistochemical analyses demonstrated that the RAS was only activated in the left myocardium, as evidenced by a decreased ACE2/ACE ratio following APE­CA and ROSC, but not the right myocardium. Ultrastructural analysis confirmed myocardial apoptosis in the left and right myocardium. Furthermore, B­cell lymphoma 2 (Bcl­2)­associated X protein (Bax) and caspase­3 levels were elevated and Bcl­2 levels were decreased in the left myocardium following APE­CA and ROSC. Treatment with the ACE inhibitor captopril for 30 min after initiation of ROSC prevented the increase in Bax and the decrease in Bcl­2 in the left myocardium compared with that in saline­treated pigs. Captopril also inhibited the activation of extracellular signal­regulated kinase (ERK)1/2 in the left myocardium. The results of the present study suggest that an imbalance in the ACE2/ACE axis has an important role in myocardial apoptosis following APE­CA, which may be attributed to decreased ERK1/2 activation. In addition, it was indicated that captopril prevents apoptosis in the left myocardium after ROSC.


Subject(s)
Apoptosis , Heart Arrest/enzymology , Heart Arrest/etiology , Myocardium/enzymology , Myocardium/pathology , Peptidyl-Dipeptidase A/metabolism , Pulmonary Embolism/complications , Acute Disease , Angiotensin-Converting Enzyme 2 , Animals , Apoptosis/drug effects , Captopril/pharmacology , Disease Models, Animal , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Myocardium/ultrastructure , Renin-Angiotensin System/drug effects , Signal Transduction/drug effects , Swine
3.
Resuscitation ; 136: 14-20, 2019 03.
Article in English | MEDLINE | ID: mdl-30654013

ABSTRACT

BACKGROUND: Neuron-specific-enolase (NSE) is frequently used to predict the neurologic outcome in persistently unconscious patients after cardiopulmonary resuscitation (CPR). However, its predictive value is unclear in the setting of veno-arterial extracorporeal membrane oxygenation therapy (ECMO). Aim of this project is to evaluate the predictive value of NSE in ECMO patients. METHODS: NSE was measured after 24, 48, and 72 h in post-CPR ECMO patients. Neurologic status was evaluated using the best Cerebral Performance Categories Score (CPC) during the hospital stay. Patients who deceased within the first 24 h and patients who were awake during the first 24 h were excluded. ROC curves were calculated to assess the discriminative ability of single NSE measurements. Trajectories of serial NSE values were investigated using latent class mixed models. RESULTS: The derivation cohort consisted of 65 patients, 30-day all-cause mortality was 47.7% and a poor neurological outcome with a CPC score of 4-5 was seen 30.7%. NSE measurement after 48 h showed the best discrimination for poor neurological outcome (AUC of 0.87 in the ROC curve; cut-off value of 70 µg/L). Specificity was highest if using serial NSE measurements at all three time points. These results could be validated in an external cohort of 64 patients. CONCLUSION: In post-CPR patients on ECMO, NSE can be used to assess the neurologic outcome. Importantly, specificity was highest if using serial NSE measurements. Further research using prospective datasets is needed to verify these findings.


Subject(s)
Extracorporeal Membrane Oxygenation/statistics & numerical data , Heart Arrest/therapy , Phosphopyruvate Hydratase/blood , Aged , Area Under Curve , Biomarkers/blood , Cardiopulmonary Resuscitation/adverse effects , Cardiopulmonary Resuscitation/statistics & numerical data , Female , Heart Arrest/enzymology , Heart Arrest/mortality , Humans , Male , Middle Aged , Predictive Value of Tests , Retrospective Studies , Time Factors
4.
J Am Heart Assoc ; 7(12)2018 06 12.
Article in English | MEDLINE | ID: mdl-29895585

ABSTRACT

BACKGROUND: Sudden cardiac arrest (CA) often results in severe injury to the brain, and neuroprotection after CA has proved to be difficult to achieve. Herein, we sought to investigate the effects of metformin pretreatment on brain injury secondary to CA and cardiopulmonary resuscitation. METHODS AND RESULTS: Rats were subjected to 9-minute asphyxial CA after receiving daily metformin treatment for 2 weeks. Survival rate, neurologic deficit scores, neuronal loss, AMP-activated protein kinase (AMPK), and autophagy activation were assessed at indicated time points within the first 7 days after return of spontaneous circulation. Our results showed that metformin pretreatment elevated the 7-day survival rate from 55% to 85% and significantly reduced neurologic deficit scores. Moreover, metformin ameliorated CA-induced neuronal degeneration and glial activation in the hippocampal CA1 region, which was accompanied by augmented AMPK phosphorylation and autophagy activation in affected neuronal tissue. Inhibition of AMPK or autophagy with pharmacological inhibitors abolished metformin-afforded neuroprotection, and augmented autophagy induction by metformin treatment appeared downstream of AMPK activation. CONCLUSIONS: Taken together, our data demonstrate, for the first time, that metformin confers neuroprotection against ischemic brain injury after CA/cardiopulmonary resuscitation by augmenting AMPK-dependent autophagy activation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy/drug effects , Brain Ischemia/prevention & control , CA1 Region, Hippocampal/drug effects , Cardiopulmonary Resuscitation/adverse effects , Heart Arrest/therapy , Metformin/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Apoptosis/drug effects , Brain Ischemia/enzymology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , CA1 Region, Hippocampal/enzymology , CA1 Region, Hippocampal/physiopathology , CA1 Region, Hippocampal/ultrastructure , Disease Models, Animal , Heart Arrest/enzymology , Heart Arrest/pathology , Heart Arrest/physiopathology , Male , Neurons/enzymology , Neurons/ultrastructure , Phosphorylation , Rats, Sprague-Dawley , Signal Transduction
5.
Shock ; 49(6): 704-711, 2018 06.
Article in English | MEDLINE | ID: mdl-28846566

ABSTRACT

No pharmacological interventions are currently available to provide neuroprotection for patients suffering from cardiac arrest. Dichloroacetate (DCA) is a pyruvate dehydrogenase kinase inhibitor, which activates pyruvate dehydrogenase (PDH), and increases cell adenosine triphosphate (ATP) production by promoting influx of pyruvate into the Krebs cycle. In this study, we investigated the effects of DCA on post-resuscitation neurological injury in an asphyxial cardiac arrest rat model. Asphyxial cardiac arrest was established by endotracheal tube clamping. A total of 111 rats were randomized into three groups: Sham group, Control group, and DCA intervention group. Animals in DCA intervention group were intraperitoneally administered DCA with a loading dose of 80 mg/kg at 15 min after return of spontaneous circulation (ROSC), whereas rats in the Control group received equivalent volume of saline. DCA treatment increased 3-day survival time, and reduced neurologic deficit scores at 24, 48, and 72 h after ROSC. It also attenuated cellular apoptosis and neuronal damage in the hippocampal cornuammonis one region by hematoxylin-eosin staining and TdT-mediated dUTP nick-end labeling assay. In addition, DCA reduced the messenger RNA expression of tumor necrosis factor α and interleukin 1ß in brain hippocampus and cortex after ROSC. Furthermore, DCA treatment significantly increased ATP production, PDH activity, and decreased blood glucose, lactate, and brain pyruvate levels after ROSC. Our results suggested that DCA has neuroprotective effects on brain injury after cardiac arrest, and its salutary effects were associated with an increase of mitochondrial energy metabolism in the brain through activation of PDH activity.


Subject(s)
Brain Injuries , Dichloroacetic Acid/pharmacology , Heart Arrest , Neuroprotective Agents/pharmacology , Pyruvate Dehydrogenase Complex/metabolism , Animals , Brain Injuries/enzymology , Brain Injuries/pathology , Brain Injuries/prevention & control , Energy Metabolism/drug effects , Enzyme Activation/drug effects , Heart Arrest/drug therapy , Heart Arrest/enzymology , Heart Arrest/pathology , Male , Oxidation-Reduction/drug effects , Rats , Rats, Sprague-Dawley , Resuscitation
6.
PLoS One ; 12(9): e0185046, 2017.
Article in English | MEDLINE | ID: mdl-28934276

ABSTRACT

RATIONALE: Post-ischemic changes in cellular metabolism alter myocardial and neurological function. Pyruvate dehydrogenase (PDH), the limiting step in mitochondrial glucose oxidation, is inhibited by increased expression of PDH kinase (PDK) during ischemia/reperfusion injury. This results in decreased utilization of glucose to generate cellular ATP. Post-cardiac arrest (CA) hypothermia improves outcomes and alters metabolism, but its influence on PDH and PDK activity following CA are unknown. We hypothesized that therapeutic hypothermia (TH) following CA is associated with the inhibition of PDK activity and increased PDH activity. We further hypothesized that an inhibitor of PDK activity, dichloroacetate (DCA), would improve PDH activity and post-CA outcomes. METHODS AND RESULTS: Anesthetized and ventilated adult female C57BL/6 wild-type mice underwent a 12-minute KCl-induced CA followed by cardiopulmonary resuscitation. Compared to normothermic (37°C) CA controls, administering TH (30°C) improved overall survival (72-hour survival rate: 62.5% vs. 28.6%, P<0.001), post-resuscitation myocardial function (ejection fraction: 50.9±3.1% vs. 27.2±2.0%, P<0.001; aorta systolic pressure: 132.7±7.3 vs. 72.3±3.0 mmHg, P<0.001), and neurological scores at 72-hour post CA (9.5±1.3 vs. 5.4±1.3, P<0.05). In both heart and brain, CA increased lactate concentrations (1.9-fold and 3.1-fold increase, respectively, P<0.01), decreased PDH enzyme activity (24% and 50% reduction, respectively, P<0.01), and increased PDK protein expressions (1.2-fold and 1.9-fold, respectively, P<0.01). In contrast, post-CA treatment with TH normalized lactate concentrations (P<0.01 and P<0.05) and PDK expressions (P<0.001 and P<0.05), while increasing PDH activity (P<0.01 and P<0.01) in both the heart and brain. Additionally, treatment with DCA (0.2 mg/g body weight) 30 min prior to CA improved both myocardial hemodynamics 2 hours post-CA (aortic systolic pressure: 123±3 vs. 96±4 mmHg, P<0.001) and 72-hour survival rates (50% vs. 19%, P<0.05) in normothermic animals. CONCLUSIONS: Enhanced PDH activity in the setting of TH or DCA administration is associated with improved post-CA resuscitation outcomes. PDH is a promising therapeutic target for improving post-CA outcomes.


Subject(s)
Dichloroacetic Acid/therapeutic use , Heart Arrest/therapy , Hypothermia, Induced , Pyruvate Dehydrogenase Complex/metabolism , Animals , Cardiopulmonary Resuscitation , Combined Modality Therapy , Female , Heart Arrest/enzymology , Heart Arrest/mortality , Hemodynamics , Mice , Mice, Inbred C57BL , Survival Rate
7.
Biomed Res Int ; 2016: 1253842, 2016.
Article in English | MEDLINE | ID: mdl-27648441

ABSTRACT

Objective. We investigated whether and how diazoxide can attenuate brain injury after cardiopulmonary resuscitation (CPR) by selective opening of mitochondrial ATP-sensitive potassium (mitoKATP) channels. Methods. Adult male Sprague-Dawley rats with induced cerebral ischemia (n = 10 per group) received an intraperitoneal injection of 0.1% dimethyl sulfoxide (1 mL; vehicle group), diazoxide (10 mg/kg; DZ group), or diazoxide (10 mg/kg) plus 5-hydroxydecanoate (5 mg/kg; DZ + 5-HD group) 30 min after CPR. The control group (sham group, n = 5) underwent sham operation, without cardiac arrest. Mitochondrial respiratory control rate (RCR) was determined. Brain cell apoptosis was assessed using TUNEL staining. Expression of Bcl-2, Bax, and protein kinase C epsilon (PKCε) in the cerebral cortex was determined by Western blotting and immunohistochemistry. Results. The neurological deficit scores (NDS) in the vehicle group decreased significantly at 24 h and 48 h after CPR. Diazoxide significantly improved NDS and mitochondrial RCR after CPR at both time points; 5-HD cotreatment abolished these effects. Diazoxide decreased TUNEL-positive cells following CPR, upregulated Bcl-2 and PKCε, downregulated Bax, and increased the Bcl-2/Bax ratio; 5-HD cotreatment reversed these effects. Conclusions. Diazoxide attenuates postresuscitation brain injury, protects mitochondrial function, inhibits brain cell apoptosis, and activates the PKC pathway by opening mitoKATP channels.


Subject(s)
Asphyxia/therapy , Brain Injuries , Diazoxide/pharmacology , Heart Arrest/therapy , Nerve Tissue Proteins/metabolism , Potassium Channels/metabolism , Resuscitation/adverse effects , Animals , Asphyxia/enzymology , Brain Injuries/enzymology , Brain Injuries/etiology , Brain Injuries/prevention & control , Cerebral Cortex/enzymology , Disease Models, Animal , Heart Arrest/enzymology , Male , Rats , Rats, Sprague-Dawley
8.
Naunyn Schmiedebergs Arch Pharmacol ; 389(11): 1159-1169, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27449068

ABSTRACT

Acute pulmonary embolism (APE) has a very high mortality rate, especially at cardiac arrest and even after the return of spontaneous circulation (ROSC). This study investigated the protective effect of the angiotensin-converting enzyme (ACE) inhibitor captopril on postresuscitation hemodynamics, in a porcine model of cardiac arrest established by APE. Twenty-nine Beijing Landrace pigs were infused with an autologous thrombus leading to cardiac arrest and subjected to standard cardiopulmonary resuscitation and thrombolysis. Ten resuscitated pigs were randomly and equally apportioned to receive either captopril (22.22 mg/kg) infusion or the same volume saline, 30 min after ROSC. Hemodynamic changes and ACE-Ang II-angiotensin II type 1 receptor (AT1R) and ACE2/Ang-(1-7)/Mas receptor axis levels were determined. APE was associated with a decline in mean arterial pressure and a dramatic increase in pulmonary artery pressure and mean right ventricular pressure. After ROSC, captopril infusion was associated with significantly lower mean right ventricular pressure and systemic and pulmonary vascular resistance, faster heart rate, and higher Ang-(1-7) levels, ACE2/ACE, and Ang-(1-7)/Ang II, compared with the saline infusion. The ACE2/Ang-(1-7)/Mas pathway correlated negatively with external vascular lung water and pulmonary vascular permeability and positively with the right cardiac index. In conclusion, in a pig model of APE leading to cardiac arrest, captopril infusion was associated with less mean right ventricular pressure overload after resuscitation, compared with saline infusion. The reduction in systemic and pulmonary vascular resistance associated with captopril may be by inhibiting the ACE-Ang II-AT1R axis and activating the ACE2/Ang-(1-7)/Mas axis.


Subject(s)
Angiotensin I/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Captopril/pharmacology , Cardiopulmonary Resuscitation , Heart Arrest/therapy , Hemodynamics/drug effects , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/metabolism , Proto-Oncogene Proteins/metabolism , Pulmonary Embolism/therapy , Receptors, G-Protein-Coupled/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Arterial Pressure/drug effects , Biomarkers/blood , Capillary Permeability/drug effects , Disease Models, Animal , Enzyme Activation , Female , Heart Arrest/blood , Heart Arrest/enzymology , Heart Arrest/physiopathology , Male , Proto-Oncogene Mas , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Pulmonary Edema/enzymology , Pulmonary Edema/physiopathology , Pulmonary Edema/prevention & control , Pulmonary Embolism/blood , Pulmonary Embolism/enzymology , Pulmonary Embolism/physiopathology , Renin-Angiotensin System/drug effects , Signal Transduction/drug effects , Sus scrofa , Thrombolytic Therapy , Time Factors , Vascular Resistance/drug effects , Ventricular Function, Right/drug effects , Ventricular Pressure/drug effects
9.
Int J Clin Exp Pathol ; 8(9): 10792-9, 2015.
Article in English | MEDLINE | ID: mdl-26617791

ABSTRACT

OBJECTIVE: This study aims to investigate the regulation effects of ulinastatin (UT1) on the expression of spermidine/spermine -N1-acetyltransferase 2 (SSAT2) and aquaporin 4 (AQP4) in myocardial tissue of rats after cardiopulmonary resuscitation (CPR) and their correlations. METHODS: A total of 90 adult SD rats were divided into sham operation group (A, n=30), model group (B, n=30) and UT1 group (C, n=30). The cardiac arrest (CA) and CPR model was established by asphyxia method. Left ventricular fractional shortening (LVFS), left ventricular ejection fraction (LVEF) and E/A peak ratio of mitral valve in three groups were collected by ultrasonic echocardiography. Apoptosis of myocardial cells was detected by DAPI staining. The expression levels of SSAT2 and AQP4 were detected by RT-PCR, Western blotting and immunohistochemical methods. RESULTS: UT1 could significantly improve the levels of LVFS, LVEF and E/A ratio and decrease myocardial cell apoptosis. As compared with group B, the expression level of SSAT2 increased and the expression level of AQP4 decreased in group C (P<0.01). SSAT2 was the most in group A and the least in group B while AQP4 was the least in group A and the most in group B (P<0.01). There was positive correlation between SSAT2 and cardiac function in CRP model while there was negative correlation between AQP4 and cardiac function (P<0.01). The expression of SSAT2 and AQP4 protein in myocardial tissue was negatively correlated in CRP model (r=-0.920, P<0.01). CONCLUSIONS: UT1 can effectively reduce the cardiac function damage caused by CRP, which could be related with the increased SSAT2 and decreased AQP4.


Subject(s)
Acetyltransferases/metabolism , Aquaporin 4/metabolism , Cardiopulmonary Resuscitation/adverse effects , Cardiotonic Agents/pharmacology , Glycoproteins/pharmacology , Heart Arrest/therapy , Myocardium/enzymology , Acetyltransferases/genetics , Animals , Apoptosis/drug effects , Aquaporin 4/genetics , Cytoprotection , Disease Models, Animal , Gene Expression Regulation , Heart Arrest/diagnostic imaging , Heart Arrest/enzymology , Heart Arrest/genetics , Heart Arrest/physiopathology , Myocardial Contraction/drug effects , Myocardium/pathology , Rats, Sprague-Dawley , Stroke Volume/drug effects , Ultrasonography , Ventricular Function, Left/drug effects
10.
Dimens Crit Care Nurs ; 33(6): 309-15, 2014.
Article in English | MEDLINE | ID: mdl-25280197

ABSTRACT

BACKGROUND: Coronary artery disease is a major problem in the United States, affecting 785 000 individuals, with the most serious event being a cardiopulmonary arrest. Families of post-cardiac arrest patients are left with difficult decisions to be made. OBJECTIVE: This article is a comprehensive literature review examining the current research available to health care professionals about the biomarker serum neuron-specific enolase (NSE) and its use in predicting neurological outcomes in comatose post-cardiac arrest patients. METHODS: Using the bibliographic databases CINAHL, Plus, EBSCOhost, MEDLINE, PubMed, Google Scholar, H. W. Wilson, Cochrane, and NEXUS, from the period 2003 to 2013, revealed there is minimal research or literature on NSE predicting neurological outcomes post-cardiac arrest. RESULTS: Research on this particular biomarker is relatively new, and more research is necessary to establish an adequate amount of support. At this time, an exact NSE cutoff value or its exact ability to help predict neurological outcomes is unable to be established, and further research is necessary. DISCUSSION: This literature review should provide a basic understanding of NSE and its ability to help predict neurological outcomes of post-cardiac arrest patients earlier. The research provides evidence that NSE should be considered in determining outcomes. However, more research is necessary before nursing practice is changed and implemented into patient care.


Subject(s)
Brain Diseases/enzymology , Coma/enzymology , Heart Arrest/enzymology , Phosphopyruvate Hydratase/blood , Biomarkers/blood , Brain Diseases/mortality , Coma/mortality , Heart Arrest/mortality , Humans , Predictive Value of Tests , Prognosis
11.
Eur J Clin Invest ; 44(10): 965-71, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25186018

ABSTRACT

OBJECTIVES: The postcardiac arrest syndrome occurs after global hypoxia leading to microcirculatory impairment. Nitric oxide (NO) is a key molecule regulating microvascular function. The enzyme arginase has been suggested to modulate microvascular function by regulating NO metabolism. Therefore, we investigated whether arginase increases following global hypoxia and resuscitation and tested whether arginase inhibition influences altered microcirculation in resuscitated patients. METHODS: To determine the effect of global hypoxia on circulating arginase levels, fourteen healthy subjects were exposed to hypoxia in a normobaric hypoxia chamber (FiO² = 9·9%). In addition, 31 resuscitated patients were characterized clinically, and arginase 1 was measured on days 1 and 3. In eight resuscitated patients, a microcirculatory analysis was performed using a sidestream darkfield microcirculation camera. Perfused capillary density (PCD) was recorded before and after sublingual incubation of N-omega-hydroxy-nor-l-arginine (nor-NOHA) alone or together with the NOS inhibitor NG-monomethyl-l-arginine (l-NMMA). RESULTS: Circulating arginase 1 levels increased in healthy volunteers following global hypoxia in the hypoxic chamber (P < 0·01). In addition, arginase 1 levels were higher on day 1 (69·1 ± 83·3 ng/mL) and on day 3 (44·2 ± 65·6 ng/mL) after resuscitation than in control subjects (P < 0·001). Incubation of the sublingual mucosa with nor-NOHA increased microcirculatory perfusion (P < 0·001). This effect was inhibited by co-incubation with K-NMMA. CONCLUSIONS: Circulating arginase 1 levels are increased following exposure to global hypoxia and in patients who have been successfully resuscitated after cardiac arrest. Topical arginase inhibition improves microcirculatory perfusion following resuscitation. This is of potential therapeutic importance for the postcardiac arrest syndrome.


Subject(s)
Arginase/metabolism , Cardiopulmonary Resuscitation , Heart Arrest/enzymology , Hypoxia/enzymology , Microcirculation/physiology , Adult , Arginine/analogs & derivatives , Arginine/pharmacology , Capillaries/physiology , Enzyme Inhibitors/pharmacology , Female , Heart Arrest/therapy , Humans , Male , Microcirculation/drug effects , Nitric Oxide/metabolism , omega-N-Methylarginine/pharmacology
12.
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
13.
Scand J Clin Lab Invest ; 74(1): 27-36, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24266783

ABSTRACT

BACKGROUND: Confined ongoing ischemia after ischemia-reperfusion injury (IRI) may alter myocardial recovery. We evaluated in a rat cardiac transplantation model whether distal persistent myocardial ischemia (dMI) and remote preconditioning (RPreC) have a remote myocardial impact after IRI. MATERIAL AND METHODS: Syngeneic heterotopic cardiac transplantation was performed on 29 Fischer344 rats to induce IRI, including nine rats which underwent distal ligation of the left anterior coronary artery (LAD) to yield distal MI (IRI+ dMI). RPreC was applied by occluding the left renal artery 5 min prior to reperfusion in six rats with IRI (IRI+ RPreC) as well as in seven with distal MI (IRI+ dMI+ RPreC). Microdialysis, histology and qRT-PCR for inducible nitric oxide synthase (iNOS) and endothelial nitric oxide synthase (eNOS) were performed after graft harvesting. RESULTS: In contrast to IRI + dMI + RPreC (39 ± 7 µmol), glutamate decreased in IRI + RPreC and IRI + dMI as compared with IRI (26 ± 3 and 31 ± 8 vs 91 ± 20, µmol respectively, p < 0.007). The relative number of vacuolated intramyocardial artery nuclei decreased in IRI + dMI as compared with IRI (0.02 ± 0.01, range 0-12 vs. 0.42 ± 0.31, range 0-3.25 PSU respectively, p < 0.04). iNOS expression decreased in IRI + RPreC as compared with IRI (p < 0.04), and eNOS expression decreased in IRI + dMI + RPreC as compared with IRI + dMI (p < 0.006) along with increased glycerol release. CONCLUSIONS: dMI after IRI has a potentially beneficial myocardial impact after cardiac arrest, which is hampered by RPreC.


Subject(s)
Heart Arrest/surgery , Ischemic Preconditioning, Myocardial , Myocardial Reperfusion Injury/prevention & control , Animals , Coronary Vessels/enzymology , Coronary Vessels/pathology , Gene Expression , Glutamic Acid/metabolism , Heart Arrest/enzymology , Heart Transplantation , Myocardial Reperfusion Injury/enzymology , Myocardium/metabolism , Myocardium/pathology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Rats , Rats, Inbred F344
14.
J Cereb Blood Flow Metab ; 33(10): 1574-81, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23820647

ABSTRACT

Cardiac arrest (CA) causes hippocampal neuronal death that frequently leads to severe loss of memory function in survivors. No specific treatment is available to reduce neuronal death and improve functional outcome. The brain's inflammatory response to ischemia can exacerbate injury and provides a potential treatment target. We hypothesized that microglia are activated by CA and contribute to neuronal loss. We used a mouse model to determine whether pharmacologic inhibition of the proinflammatory microglial enzyme soluble epoxide hydrolase (sEH) after CA alters microglial activation and neuronal death. The sEH inhibitor 4-phenylchalcone oxide (4-PCO) was administered after successful cardiopulmonary resuscitation (CPR). The 4-PCO treatment significantly reduced neuronal death and improved memory function after CA/CPR. We found early activation of microglia and increased expression of inflammatory tumor necrosis factor (TNF)-α and interleukin (IL)-1ß in the hippocampus after CA/CPR, which was unchanged after 4-PCO treatment, while expression of antiinflammatory IL-10 increased significantly. We conclude that sEH inhibition after CA/CPR can alter the transcription profile in activated microglia to selectively induce antiinflammatory and neuroprotective IL-10 and reduce subsequent neuronal death. Switching microglial gene expression toward a neuroprotective phenotype is a promising new therapeutic approach for ischemic brain injury.


Subject(s)
Cardiopulmonary Resuscitation , Epoxide Hydrolases/antagonists & inhibitors , Heart Arrest/drug therapy , Heart Arrest/pathology , Microglia/pathology , Neurons/pathology , Animals , Cell Death/drug effects , Cell Survival/drug effects , Chalcones/administration & dosage , Chalcones/therapeutic use , Heart Arrest/enzymology , Heart Arrest/immunology , Interleukin-10/biosynthesis , Interleukin-1beta/biosynthesis , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Neurons/drug effects , Neurons/immunology , Tumor Necrosis Factor-alpha/biosynthesis
15.
Scand Cardiovasc J Suppl ; 47(1): 58-64, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22994795

ABSTRACT

OBJECTIVES: Resuscitation after cardiac arrest may lead to ischemia-reperfusion injury and infarction. We evaluated whether sildenafil, a phosphodiesterase-5 inhibitor, has an impact on recovery after cardiac arrest in a rat cardiac transplantation model. DESIGN: Sixty-one Fischer344 rats underwent syngeneic heterotopic cardiac transplantation after ischemia and ligation of the left anterior coronary artery of the heart to yield myocardial infarction (IRI + MI). Of these, 22 rats received subcutaneously injected sildenafil (1 mg/kg/day) (IRI +MI + S). Twenty-three additional grafted animals with transplantation only served as controls with ischemia reperfusion injury (IRI). After 2 days, immunohistochemistry for eNOS, and RT-PCR for iNOS and Aquaporin-7 were performed after graft harvesting and histology. RESULTS: Two days after transplantation, remote intramyocardial arteries were more preserved in IRI + MI + S as compared with IRI +MI and IRI (0.74 ± 0.14, 0.56 ± 0.23 and 0.55 ± 0.22, PSU, p < 0.05, respectively). Decreased eNOS staining confirmed the presence of developing infarction in IRI + MI and IRI + MI + S. The expression of iNOS was significantly lower during IRI + MI +S as compared with IRI + MI (0.02 ± 0.01 and 1.02 ± 0.02, FC, p < 0.05). CONCLUSIONS: Administered at the onset of reperfusion and developing infarction, sildenafil has an impact on myocardial recovery after cardiac arrest and ischemia.


Subject(s)
Heart Arrest/drug therapy , Myocardial Infarction/complications , Myocardial Reperfusion Injury/prevention & control , Phosphodiesterase 5 Inhibitors/administration & dosage , Piperazines/administration & dosage , Sulfones/administration & dosage , Vasodilator Agents/administration & dosage , Animals , Aquaporins/genetics , Coronary Vessels/drug effects , Coronary Vessels/enzymology , Coronary Vessels/pathology , Disease Models, Animal , Drug Administration Schedule , Heart Arrest/enzymology , Heart Arrest/etiology , Heart Arrest/genetics , Heart Arrest/pathology , Heart Transplantation , Immunohistochemistry , Injections, Subcutaneous , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardium/enzymology , Myocardium/pathology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Purines/administration & dosage , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Sildenafil Citrate , Time Factors
16.
Cardiovasc Toxicol ; 12(4): 350-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22814936

ABSTRACT

Ischemia/reperfusion damage is common during open-heart surgery. Activation of aldehyde dehydrogenase-2 can significantly reduce ischemia/reperfusion damage. We hypothesized that adding aldehyde dehydrogenase-2 agonist to regular cardioplegia solution would further ameliorate ischemia/reperfusion damage. Alda-1 was used as an aldehyde dehydrogenase-2 agonist. Cardioprotection by histidine-tryptophan-ketoglutarate solution with and without Alda-1 was compared using an ex vivo perfused rat heart model of ischemia/reperfusion. Three groups of ex vivo rat hearts endured different treatments with variant ischemia or an ischemia/reperfusion time course: sham, no ischemia/reperfusion; histidine-tryptophan-ketoglutarate; and histidine-tryptophan-ketoglutarate plus Alda-1. Aldehyde dehydrogenase-2 expressions and activities, oxidative parameters (including 4-hydroxy-2-nonenal-His adducts, malondialdehyde levels, and glutathione/oxidized glutathione ratios), myocardial protein carbonyl levels, coronary effluents creatine kinase isoenzyme MB levels, and heart function parameters were measured and compared. Alda-1 significantly elevated myocardium aldehyde dehydrogenase-2 activity (P < .01). Increased aldehyde dehydrogenase-2 activity in turn attenuated ischemia/reperfusion-induced elevation in cardiac aldehydes, creatine kinase isoenzyme MB leakage, and protein carbonyl formation (P < .01). The Alda-1 group also obtained higher glutathione/oxidized glutathione ratios (P < .01). Aldehyde dehydrogenase-2 activation alleviated ischemia/reperfusion-induced cardiomyocyte contractile function impairment as evidenced by improved maximal velocity of pressure development and decline, left ventricular developed pressure, and heart rate (P < .01). Alda-1 supplementation can significantly improve the cardioprotection effect of cardioplegia solution, possibly through activation of aldehyde dehydrogenase-2, to remove toxic aldehydes. This may aid in the identification of novel cardioplegia solutions.


Subject(s)
Aldehyde Dehydrogenase/physiology , Cardiotonic Agents/therapeutic use , Heart Arrest, Induced/methods , Heart Arrest/enzymology , Mitochondrial Proteins/physiology , Myocardial Reperfusion Injury/enzymology , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase, Mitochondrial , Animals , Benzamides/pharmacology , Benzamides/therapeutic use , Benzodioxoles/pharmacology , Benzodioxoles/therapeutic use , Cardioplegic Solutions/pharmacology , Cardioplegic Solutions/therapeutic use , Cardiotonic Agents/agonists , Enzyme Activation/physiology , Heart Arrest/prevention & control , Male , Mitochondrial Proteins/agonists , Mitochondrial Proteins/metabolism , Myocardial Reperfusion Injury/prevention & control , Rats , Rats, Wistar , Up-Regulation/drug effects , Up-Regulation/physiology
17.
Resuscitation ; 83(2): 197-201, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21854733

ABSTRACT

AIM: To study the systemic levels of matrix metalloproteinases (MMP) -7, -8 and -9 and their inhibitor TIMP-1 in cardiac arrest patients and the association with mild therapeutic hypothermia treatment on the serum concentration of these enzymes. METHODS: MMP-7, -8 and -9 and tissue inhibitor of metalloproteinases-1 (TIMP-1) were analysed in blood samples obtained from 51 patients resuscitated from cardiac arrest. The samples were taken at 24 and 48 h from restoration of spontaneous circulation (ROSC). The biomarker levels were compared between patients (N=51) and healthy controls (N=10) and between patients who did (N=30) and patients who did not (N=21) receive mild therapeutic hypothermia. RESULTS: MMP-7 (median 0.47 ng/ml), MMP-8 (median 31.16 ng/ml) and MMP-9 (median 253.00 ng/ml) levels were elevated and TIMP-1 levels suppressed (median 78.50 ng/ml) in cardiac arrest patients as compared with healthy controls at 24h from ROSC. Hypothermia treatment associated with attenuated elevation of MMP-9 (p=0.001) but not MMP-8 (p=0.02) or MMP-7 (p=0.69). Concentrations of MMPs -7, -8 and -9 correlated with the leukocyte count but not with C-reactive protein (CRP) or neurone-specific enolase (NSE) levels. CONCLUSION: We demonstrated that the systemic levels of MMP-7, -8 and -9 but not TIMP-1 are elevated in cardiac arrest patients in the 48 h post-resuscitation period relative to the healthy controls. Patients who received therapeutic hypothermia had lower MMP-9 levels compared to non-hypothermia treated patients, which generates hypothesis about attenuation of inflammatory response by hypothermia treatment.


Subject(s)
Cardiopulmonary Resuscitation/methods , Heart Arrest/enzymology , Hypothermia, Induced , Matrix Metalloproteinases/blood , Aged , Arrhythmias, Cardiac/complications , Biomarkers/blood , Enzyme-Linked Immunosorbent Assay , Female , Heart Arrest/etiology , Heart Arrest/therapy , Humans , Male , Middle Aged , Treatment Outcome
18.
Drug Metab Dispos ; 39(12): 2209-18, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21868471

ABSTRACT

Mild therapeutic hypothermia is emerging clinically as a neuroprotection therapy for individuals experiencing cardiac arrest (CA); however, its effects combined with disease pathogenesis on drug disposition and response have not been fully elucidated. We determined the activities of four major hepatic-metabolizing enzymes (CYP3A, CYP2C, CYP2D, and CYP2E) during hypothermia after experimental CA in rats by evaluating the pharmacokinetics of their probe drugs as a function of altered body temperature. Animals were randomized into sham normothermia (37.5-38°C), CA normothermia, sham hypothermia (32.5-33°C), and CA hypothermia groups. Probe drugs (midazolam, diclofenac, dextromethorphan, and chlorzoxazone) were given simultaneously by intravenous bolus after temperature stabilization. Multiple blood samples were collected between 0 and 8 h after drug administration. Pharmacokinetic (PK) analysis was conducted using a noncompartmental approach and population PK modeling. Noncompartmental analysis showed that the clearance of midazolam (CYP3A) in CA hypothermia was reduced from sham normothermia rats (681.6 ± 190.0 versus 1268.8 ± 348.9 ml · h(-1) · kg(-1), p < 0.05). The clearance of chlorzoxazone (CYP2E) in CA hypothermia was also reduced from sham normothermia rats (229.6 ± 75.6 versus 561.89 ± 215.9 ml · h(-1) · kg(-1), p < 0.05). Population PK analysis further demonstrated the decreased clearance of midazolam (CYP3A) was associated with CA injury (p < 0.05). The decreased clearance of chlorzoxazone (CYP2E1) was also associated with CA injury (p < 0.01). Hypothermia was found to be associated with the decreased volume of distribution of midazolam (V(1)), dextromethorphan (V(1)), and peripheral compartment for chlorzoxazone (V(2)) (p < 0.05, p < 0.05, and p < 0.01, respectively). Our data indicate that hypothermia, CA, and their interaction alter cytochrome P450-isoform specific activities in an isoform-specific manner.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Heart Arrest/therapy , Hypothermia, Induced , Isoenzymes/metabolism , Animals , Body Temperature , Chromatography, Liquid , Heart Arrest/enzymology , Male , Mass Spectrometry , Pharmacokinetics , Rats , Rats, Sprague-Dawley
19.
BMC Cardiovasc Disord ; 11: 48, 2011 Aug 08.
Article in English | MEDLINE | ID: mdl-21824428

ABSTRACT

BACKGROUND: The prediction of neurological outcome in comatose patients after cardiac arrest has major ethical and socioeconomic implications. The purpose of this study was to assess the capability of serum neuron-specific enolase (NSE), a biomarker of hypoxic brain damage, to predict death or vegetative state in comatose cardiac-arrest survivors. METHODS: We conducted a prospective observational cohort study in one university hospital and one general hospital Intensive Care Unit (ICU). All consecutive patients who suffered cardiac arrest and were subsequently admitted from June 2007 to February 2009 were considered for inclusion in the study. Patients who died or awoke within the first 48 hours of admission were excluded from the analysis. Patients were followed for 3 months or until death after cardiopulmonary resuscitation. The Cerebral Performance Categories scale (CPC) was used as the outcome measure; a CPC of 4-5 was regarded as a poor outcome, and a CPC of 1-3 a good outcome. Measurement of serum NSE was performed at 24 h and at 72 h after the time of cardiac arrest using an enzyme immunoassay. Clinicians were blinded to NSE results. RESULTS: Ninety-seven patients were included. All patients were actively supported during the first days following cardiac arrest. Sixty-five patients (67%) underwent cooling after resuscitation. At 3 months 72 (74%) patients had a poor outcome (CPC 4-5) and 25 (26%) a good outcome (CPC 1-3). The median and Interquartile Range [IQR] levels of NSE at 24 h and at 72 h were significantly higher in patients with poor outcomes: NSE at 24 h: 59.4 ng/mL [37-106] versus 28.8 ng/mL [18-41] (p < 0.0001); and NSE at 72 h: 129.5 ng/mL [40-247] versus 15.7 ng/mL [12-19] (p < 0.0001). The Receiver Operator Characteristics (ROC) curve for poor outcome for the highest observed NSE value for each patient determined a cut-off value for NSE of 97 ng/mL to predict a poor neurological outcome with a specificity of 100% [95% CI = 87-100] and a sensitivity of 49% [95% CI = 37-60]. However, an approach based on a combination of SSEPs, NSE and clinical-EEG tests allowed to increase the number of patients (63/72 (88%)) identified as having a poor outcome and for whom intensive treatment could be regarded as futile. CONCLUSION: NSE levels measured early in the course of patient care for those who remained comatose after cardiac arrest were significantly higher in patients with outcomes of death or vegetative state. In addition, we provide a cut-off value for NSE (> 97 ng/mL) with 100% positive predictive value of poor outcome. Nevertheless, for decisions concerning the continuation of treatment in this setting, we emphasize that an approach based on a combination of SSEPs, NSE and clinical EEG would be more accurate for identifying patients with a poor neurological outcome.


Subject(s)
Coma/diagnosis , Coma/enzymology , Heart Arrest/diagnosis , Heart Arrest/enzymology , Phosphopyruvate Hydratase/blood , Adult , Aged , Biomarkers/blood , Cohort Studies , Coma/mortality , Female , Heart Arrest/mortality , Humans , Male , Middle Aged , Predictive Value of Tests , Prospective Studies , Survival Rate/trends , Treatment Outcome
20.
Resuscitation ; 82(1): 115-21, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20951489

ABSTRACT

STUDY AIMS: Cardiac arrest mortality is significantly affected by failure to obtain return of spontaneous circulation (ROSC) despite cardiopulmonary resuscitation (CPR). Severe myocardial dysfunction and cardiovascular collapse further affects mortality within hours of initial ROSC. Recent work suggests that enhancement of nitric oxide (NO) signaling within minutes of CPR can improve myocardial function and survival. We studied the role of NO signaling on cardiovascular outcomes following cardiac arrest and resuscitation using endothelial NO synthase knockout (NOS3(-/-)) mice. METHODS: Adult female wild-type (WT) and NOS3(-/-) mice were anesthetized, intubated, and instrumented with left-ventricular pressure-volume catheters. Cardiac arrest was induced with intravenous potassium chloride. CPR was performed after 8min of untreated arrest. ROSC rate, cardiac function, whole-blood nitrosylhemoglobin (HbNO) concentrations, heart NOS3 content and phosphorylation (p-NOS3), cyclic guanosine monophosphate (cGMP), and phospho-troponin I (p-TnI) were measured. RESULTS: Despite equal quality CPR, NOS3(-/-) mice displayed lower rates of ROSC compared to WT (47.6% [10/21] vs. 82.4% [14/17], p<0.005). Among ROSC animals, NOS3(-/-) vs. WT mice exhibited increased left-ventricular dysfunction and 120min mortality. Prior to ROSC, myocardial effectors of NO signaling including cGMP and p-TnI were decreased in NOS3(-/-) vs. WT mice (p<0.05). Following ROSC in WT mice, significant NOS3-dependent increases in circulating HbNO were seen by 120min. Significant increases in cardiac p-NOS3 occurred between end-arrest and 15min post-ROSC, while total NOS3 content was increased by 120min post-ROSC (p<0.05). CONCLUSIONS: Genetic deletion of NOS3 decreases ROSC rate and worsens post-ROSC left-ventricular function. Poor cardiovascular outcomes are associated with differences in NOS3-dependent myocardial cGMP signaling and circulating NO metabolites.


Subject(s)
DNA/genetics , Gene Deletion , Heart Arrest/genetics , Nitric Oxide Synthase Type III/genetics , Ventricular Dysfunction, Left/genetics , Animals , Blotting, Western , Cardiopulmonary Resuscitation/methods , Disease Models, Animal , Female , Heart Arrest/enzymology , Heart Arrest/therapy , Mice , Mice, Inbred C57BL , Myocardium/enzymology , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...