Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Oxid Med Cell Longev ; 2022: 7736416, 2022.
Article in English | MEDLINE | ID: mdl-35847595

ABSTRACT

Ketone bodies including ß-hydroxybutyrate (ß-HB) have been proved the therapeutic potential in diverse neurological disorders. However, the role of ß-HB in the regulation of neurological injury after cardiac arrest (CA) remains unclear. We investigated the effect of ß-HB on brain mitochondrial dysfunction and neurological function after CA. A rat model of CA was established by asphyxia. The rats were randomly divided into three groups: sham group, control group, and ß-HB group. Animals received 200 mg/kg ß-HB or same volume vehicle at 10 minutes after return of spontaneous circulation by intraperitoneal injection. Neurological function was evaluated by neurologic deficit score and Y-maze. Neuronal cell loss and apoptosis were detected through hematoxylin-eosin staining, Nissl staining, and TdT-mediated dUTP nick-end labeling assay. Oxidative stress levels were determined by immunohistochemical staining of 4-hydoxynonenal and 8-hydroxy-2'-deoxyguanosine. Furthermore, mitochondrial ultrastructure of brain cells was observed by transmission electron microscopy. In addition, the protein expression levels of Bak, caspase 3, gasdermin D, caspase 1, brain-derived neurotrophic factor, dynamin-related protein 1 (Drp1), and phospho-Drp1 (ser616) were measured. We found that neurological function and survival rate were significantly higher in the ß-HB group compared with the control group. ß-HB also reduced neurons death and neurological oxidative stress after CA. Moreover, ß-HB reduced neurological injury from apoptosis and pyroptosis after CA. In addition, ß-HB maintained the structural integrity of brain mitochondria, prevented mitochondrial fission, and increased brain energy metabolism after CA. In conclusion, ß-HB beneficially affected the neurological function of rats after global cerebral ischemia, associated with decreased mitochondrial fission, and improved mitochondrial function. Our results suggest that ß-HB might benefit patients suffering from neurological dysfunction after CA.


Subject(s)
Heart Arrest , Mitochondrial Dynamics , Animals , Apoptosis , Ketone Bodies/metabolism , Ketone Bodies/pharmacology , Ketone Bodies/therapeutic use , Mitochondria/metabolism , Rats , Rats, Sprague-Dawley
2.
Shock ; 57(4): 576-582, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34731097

ABSTRACT

OBJECTIVES: To investigate the activity of key rate-limiting enzymes of glucose metabolism after restoration of spontaneous circulation (ROSC), to explore the potential pathophysiological mechanism of impaired myocardial energy metabolism after cardiopulmonary resuscitation (CPR). METHODS: Twenty-one male Sprague-Dawley rats were randomized into three experimental groups assigned in accordance with different observation times after ROSC: Sham, instrumented rats without induced cardiac arrest or resuscitation; post-resuscitation (PR2 h); PR24 h. In these groups, CPR, including precordial compressions and synchronized mechanical ventilation, was initiated 6 min after asphyxia-induced cardiac arrest. Hearts were harvested after ROSC and samples were used to detect high-energy phosphate and glucose metabolic enzyme activity. RESULTS: Compared with sham, the contents of phosphocreatine and adenosine triphosphate reduced in the PR2 h group, while remained unchanged in the PR24 h group. Activities of hexokinase and pyruvate kinase did not change after ROSC. Phosphofructokinase activity decreased only in the PR24 h group. Activities of pyruvate dehydrogenase and citrate synthase fell in PR2 h group and recovered in the PR24 h group. However, isocitrate dehydrogenase and α-ketoglutarate dehydrogenase activities fell in the PR2 h group, but did not recover in the PR24 h group. CONCLUSIONS: Lowered key rate-limiting enzymes activity in glucose metabolism resulted in impairment of energy production in the early stage of ROSC, but partially recovered in 24 h. This process has a role in the mechanism of impaired myocardial energy metabolism after CPR. This investigation might shed light on new strategies to treat post resuscitation myocardial dysfunction.


Subject(s)
Cardiopulmonary Resuscitation , Heart Arrest , Animals , Male , Rats , Asphyxia , Cardiopulmonary Resuscitation/methods , Glucose , Rats, Sprague-Dawley
3.
Front Pharmacol ; 12: 758080, 2021.
Article in English | MEDLINE | ID: mdl-34712142

ABSTRACT

Empagliflozin is a newly developed antidiabetic drug to reduce hyperglycaemia by highly selective inhibition of sodium-glucose co-transporter 2. Hyperglycaemia is commonly seen in patients after cardiac arrest (CA) and is associated with worse outcomes. In this study, we examined the effects of empagliflozin on cardiac function in rats with myocardial dysfunction after CA. Non-diabetic male Sprague-Dawley rats underwent ventricular fibrillation to induce CA, or sham surgery. Rats received 10 mg/kg of empagliflozin or vehicle at 10 min after return of spontaneous circulation by intraperitoneal injection. Cardiac function was assessed by echocardiography, histological analysis, molecular markers of myocardial injury, oxidative stress, mitochondrial ultrastructural integrity and metabolism. We found that empagliflozin did not influence heart rate and blood pressure, but left ventricular function and survival time were significantly higher in the empagliflozin treated group compared to the group treated with vehicle. Empagliflozin also reduced myocardial fibrosis, serum cardiac troponin I levels and myocardial oxidative stress after CA. Moreover, empagliflozin maintained the structural integrity of myocardial mitochondria and increased mitochondrial activity after CA. In addition, empagliflozin increased circulating and myocardial ketone levels as well as heart ß-hydroxy butyrate dehydrogenase 1 protein expression. Together, these metabolic changes were associated with an increase in cardiac energy metabolism. Therefore, empagliflozin favorably affected cardiac function in non-diabetic rats with acute myocardial dysfunction after CA, associated with reducing glucose levels and increasing ketone body oxidized metabolism. Our data suggest that empagliflozin might benefit patients with myocardial dysfunction after CA.

4.
Ann Transl Med ; 9(1): 8, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33553301

ABSTRACT

BACKGROUND: Dietary restriction (DR) is a well-known intervention that increases lifespan and resistance to multiple forms of acute stress, including ischemia reperfusion injury. However, the effect of DR on neurological injury after cardiac arrest (CA) remains unknown. METHODS: The effect of short-term DR (one week of 70% reduced daily diet) on neurological injury was investigated in rats using an asphyxial CA model. The survival curve was obtained using Kaplan-Meier survival analysis. Serum S-100ß levels were detected by enzyme linked immunosorbent assay. Cellular apoptosis and neuronal damage were assessed by terminal deoxyribonucleotide transferase dUTP nick end labeling assay and Nissl staining. The oxidative stress was evaluated by immunohistochemical staining of 8-hydroxy-2'-deoxyguanosine (8-OHdG). Mitochondrial biogenesis was examined by electron microscopy and mitochondrial DNA copy number determination. The protein expression was detected by western blot. The reactive oxygen species (ROS) and metabolite levels were measured by corresponding test kits. RESULTS: Short-term DR significantly improved 3-day survival, neurologic deficit scores (NDS) and decreased serum S-100ß levels after CA. Short-term DR also significantly attenuated cellular apoptosis, neuronal damage and oxidative stress in the brain after CA. In addition, short-term DR increased mitochondrial biogenesis as well as brain PGC-1α and SIRT1 protein expression after CA. Moreover, short-term DR increased adenosine triphosphate, ß-hydroxybutyrate, acetyl-CoA levels and nicotinamide adenine dinucleotide (NAD+)/reduced form of NAD+ (NADH) ratios as well as decreased serum lactate levels. CONCLUSIONS: Reduction of oxidative stress, upregulation of mitochondrial biogenesis and increase of ketone body metabolism may play a crucial role in preserving neuronal function after CA under short-term DR.

5.
Am J Emerg Med ; 38(1): 65-72, 2020 01.
Article in English | MEDLINE | ID: mdl-31027936

ABSTRACT

PURPOSE: Previous studies implicate that the mitochondrial injury may play an important role in the development of post-resuscitation myocardial dysfunction, however few of them are available regarding the ultrastructural alterations of myocardial mitochondria, mitochondrial energy producing and utilization ability in the stage of arrest time (no-low) and resuscitation time (low-flow). This study aimed to observe the dynamic changes of myocardial mitochondrial function and metabolic disorders during cardiac arrest (CA) and following cardiopulmonary resuscitation (CPR). METHODS: A total of 30 healthy male Sprague-Dawley rats were randomized into three groups: 1) VF/CPR: Ventricular fibrillation (VF) was electrically induced, and 5 min of CPR was performed after 10 min of untreated VF; 2) Untreated VF: VF was induced and untreated for 15 min; and 3) Sham: Rats were identically prepared without VF/CPR. Amplitude spectrum area (AMSA) at VF 5, 10 and 15 min were calculated from ECG signals. The rats' hearts were quickly removed at the predetermined time of 15 min after beginning the procedure to gather measurements of myocardial mitochondrial function, high-energy phosphate stores, lactate, mitochondrial ultrastructure, and myocardial glycogen. RESULTS: The mitochondrial respiratory control ratios significantly decreased after CA compared to sham group. CPR significantly increased respiratory control ratios compared with untreated VF animals. A significant decrease of myocardial glycogen was observed after CA, and a more rapid depletion of myocardial glycogen was observed in CPR animals. CPR significantly reduced the tissue lactate. The mitochondrial ultrastructure abnormalities in CPR animals were less severe than untreated VF animals. AMSA decayed during untreated VF; however, it was significantly greater in CPR group than the untreated VF group. In addition, AMSA was clearly positively correlated with ATP, but negatively correlated with myocardial glycogen. CONCLUSION: Impairment of myocardial mitochondrial function and the incapability of utilizing glycogen were observed after CA. Furthermore, optimal CPR might, in part, preserved mitochondrial function and enhanced utilization of myocardial glycogen.


Subject(s)
Cardiopulmonary Resuscitation , Heart Arrest/physiopathology , Heart Arrest/therapy , Mitochondria, Heart/physiology , Animals , Disease Models, Animal , Electrocardiography , Energy Metabolism , Glycogen/metabolism , Heart Arrest/metabolism , Heart Arrest/pathology , Lactic Acid/metabolism , Male , Mitochondria, Heart/ultrastructure , Phosphates/metabolism , Rats, Sprague-Dawley
6.
J Inflamm (Lond) ; 16: 3, 2019.
Article in English | MEDLINE | ID: mdl-30820191

ABSTRACT

BACKGROUND: Sepsis is a systemic inflammatory response syndrome caused by severe infections. LDK378, a second-generation ALK inhibitor, exhibits a potential anti-inflammatory function against sepsis. Micro- and macro-circulatory dysfunctions are pivotal elements of the pathogenesis of severe sepsis and septic shock. We hypothesized that LDK378 can improve micro- and macro-circulation of septic rats, therefore improving the outcome of survival via blocking the ALK-STING pathway to attenuate inflammatory injuries. METHODS: A septic rat model was established by the cecal ligation and puncture (CLP) method. A total of 60 rats were randomized into three groups: a sham group, CLP group, and CLP + LDK378 group (n = 20 in each group). Five rats were randomly selected from each group for the mechanism study; the remaining 15 rats in each group were involved in a survival curve examination. A sidestream dark field video microscope was used to record sublingual microcirculation and mean arterial pressure (MAP) and levels of inflammatory cytokine secretion were examined at 6 h, 30 h, and 54 h after CLP surgery. Expressions of TANK binding kinase 1 (TBK1) and its downstream targets were determined, and histological alterations to the heart, lungs, and kidneys were examined at 54 h after CLP surgery. RESULTS: We found the group that received LDK378 treatment showed increased MAP levels compared to the CLP group at 30 h and 54 h. Meanwhile, LDK378 ameliorated the perfused small vessel density and microvascular flow index, decreased the expression of TNF-a and IL-6, and upregulated the expression of IL-10 in comparison with the CLP group. LDK378 injections also downregulated the expression of TBK1 and its downstream targets. Furthermore, LDK378 treatment significantly reduced sepsis-induced organ injuries, therefore improving survival rates. CONCLUSIONS: These findings demonstrate that LDK378 treatment can improve microcirculation and reduce organ injuries in CLP-induced septic rats via the regulation of inflammatory cytokine secretion and the downstream signaling components of the ALK-STING pathway.

7.
Shock ; 51(4): 519-525, 2019 04.
Article in English | MEDLINE | ID: mdl-30875342

ABSTRACT

Cecal ligation and puncture (CLP) was used to cause severe sepsis in male Sprague-Dawley rats. There are four groups in this study: sham (n = 5), CLP (n = 10), end-tidal carbon dioxide (ETCO2) (n = 10), and mean arterial pressure (MAP) (n = 10). In ETCO2 group, fluid resuscitation (FR) began when ETCO2 at most 25 mmHg. In MAP group, FR began when MAP at most 100 mmHg. Electrocardiogram, aortic pressure, core temperature, and ETCO2 values were recorded at baseline, 2, 4, 6, 8, 10, and 12 h post-CLP. Lactate level, cardiac output (CO), perfused small vessel density (PSVD), and microvascular flow index (MFI) were assessed at the same time points as above. The results showed that MAP, CO, and ETCO2 gradually decreased after CLP. After FR, MAP, ETCO2, and CO in the ETCO2 group increased compared with the MAP group 12 h after CLP (all P < 0.05). Lactate level remains high in MAP group while decreasing in the ETCO2 group 8 h post-CLP. Both PSVD and MFI deteriorated after CLP in CLP group, though significantly improved in the ETCO2 group 8 h post-CLP. The average survival time in the ETCO2 group was significantly greater than MAP group (14.95 ±â€Š3.90 h vs. 11.15 ±â€Š1.76 h; t = 2.804, P = 0.012). Moreover, ETCO2 showed a negative correlation with lactic acid levels and a positive correlation with CO, PSVD, and MFI. In conclusion, ETCO2 can guide FR implement and improve outcomes of severe sepsis in CLP-inducted rat model. ETCO2 might be a potential index to guiding early FR in severe sepsis.


Subject(s)
Cecum/injuries , Ligation/adverse effects , Sepsis/therapy , Animals , Carbon Dioxide , Cardiac Output/physiology , Disease Models, Animal , Hemodynamics/physiology , Male , Microcirculation/physiology , Punctures/adverse effects , Rats , Rats, Sprague-Dawley , Resuscitation
8.
Shock ; 51(1): 105-113, 2019 01.
Article in English | MEDLINE | ID: mdl-29424796

ABSTRACT

BACKGROUND: Global cerebral ischemic/reperfusion (I/R) injury after cardiac arrest (CA) is a major cause of mortality and morbidity in survivors of resuscitation. We utilized a rat model of asphyxia CA to explore the functional effects and mechanisms of Sigma-1 receptor (Sig-1R) activation in cerebral protection using the Sig-1R agonist cutamesine (SA-4503). METHODS: After resuscitation, the surviving rats were randomly divided into three groups (n = 18 each): the cardiopulmonary resuscitation (CPR) group (0.9% saline at 1 mL/kg); the SA4503 low-dose group (1 mg/kg SA4503); and the SA4503 high-dose group (2.5 mg/kg SA4503). The neurological deficit scores were recorded, and the cerebral cortex was harvested for western blotting. Mitochondrial transmembrane potential, adenosine triphosphate (ATP) concentrations, calcium homeostasis, and mitochondrial ultrastructure were also studied. RESULTS: The SA4503 treatment groups exhibited improved neurological outcomes compared with the CPR group. The protein levels of caspase-3 and the endoplasmic reticulum stress markers C/EBP homologous protein and caspase-12 were lower in the SA4503 treatment groups compared with the CPR group. SA4503 treatment also normalized mitochondrial membrane potential, tissue ATP concentrations, intracellular Ca overload, and upregulated Sig-1R protein level compared with the CPR group. The SA4503 high dose treatment showed significant cerebral protective effects compared with the SA4503 low dose treatment. The therapeutic effect of SA4503 was dose-dependent. CONCLUSIONS: CA downregulated Sig-1R protein expression. Activating Sig-1R using SA4503 protected against global cerebral I/R injury in a rat model of asphyxia CA by alleviating endoplasmic reticulum stress and mitochondrial dysfunction and eventually inhibiting neuronal apoptosis.


Subject(s)
Apoptosis , Asphyxia , Endoplasmic Reticulum Stress , Heart Arrest , Neurons , Piperazines , Receptors, sigma , Resuscitation , Animals , Male , Rats , Apoptosis/drug effects , Asphyxia/metabolism , Asphyxia/pathology , Asphyxia/therapy , Brain/metabolism , Brain/pathology , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Heart Arrest/metabolism , Heart Arrest/pathology , Heart Arrest/therapy , Neurons/metabolism , Neurons/pathology , Piperazines/pharmacology , Rats, Sprague-Dawley , Receptors, sigma/agonists , Receptors, sigma/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/therapy , Sigma-1 Receptor
9.
Int J Chron Obstruct Pulmon Dis ; 13: 2351-2360, 2018.
Article in English | MEDLINE | ID: mdl-30122915

ABSTRACT

Background and aim: It is desirable to facilitate the use of an affordable, reliable, and portable spirometer, for earlier diagnosis of COPD in China, particularly in rural areas. The aim of this study was to assess the agreement of a handheld "disposable pneumotachograph" (D-PNEU) spirometer with the gold standard spirometer and to evaluate its diagnostic accuracy of spirometric classification of airflow obstruction. Subjects and methods: A total of 241 adult Chinese subjects ranging from healthy to those with mixed levels of pulmonary disease performed spirometry in a conventional body plethysmograph, and using a D-PNEU device in randomized order. The three best spirometric tests were recorded for comparative analysis. A Bland-Altman graph was created to assess the agreement between devices. Using FEV1/FVC <70% as the "gold standard" for obstruction, the accuracy of classifying the severity of airway obstruction for all subjects was assessed. For the specific individuals (n=159) able to exhale for at least 6 seconds, the accuracy of classifying airway obstruction was further assessed. For this purpose, a receiver operating characteristic curve was used to determine an optimal cutoff point of FEV1/FEV6 ratio obtained by the D-PNEU device, which matched the global definition of FEV1/FVC <70% by the traditional spirometer. Results: The Bland-Altman analysis showed that the between-device agreement for key airflow metrics was within clinically acceptable limits. The D-PNEU device had 87.1% accuracy in the classification of severity of obstruction in all 241 subjects, when using FEV1/FVC<70% as the "gold standard" for both devices. The D-PNEU device had 93.7% accuracy in the 159 individuals able to exhale for at least 6 seconds, when a cutoff point of FEV1/FEV6 was 74%. Conclusion: A disposable handheld spirometry device is capable of accurately identifying and quantifying airway obstruction in patients deemed to be at risk, however, caution should be exercised and all available brands should be tested.


Subject(s)
Airway Obstruction/diagnostic imaging , Pulmonary Disease, Chronic Obstructive/diagnosis , Spirometry/instrumentation , Adult , Airway Obstruction/physiopathology , China , Disposable Equipment , Forced Expiratory Volume , Humans , Plethysmography/instrumentation , ROC Curve , Random Allocation , Sensitivity and Specificity , Spirometry/standards , Vital Capacity
10.
Transl Res ; 194: 68-78, 2018 04.
Article in English | MEDLINE | ID: mdl-29351829

ABSTRACT

Dynamin-related protein 1 (Drp1) regulates mitochondrial fission, it has been proven that inhibition of Drp1 by mdivi-1 improves survival and attenuates cerebral ischemic injury after cardiac arrest. In this study, we compared the effects of Drp1 inhibition with therapeutic hypothermia on post-resuscitation neurologic injury in a rat model of cardiac arrest. Rats were randomized into 4 groups: mdivi-1 treatment group (n = 39), hypothermic group (n = 38), normothermic group (n = 41), and sham group (n = 12). The rats in the mdivi-1 treatment group were received intravenously 1.2 mg/kg of mdivi-1 at 1 minute after the return of spontaneous circulation (ROSC). In rats in hypothermia group, rapid cooling was initiated at 5 minutes after resuscitation, and the core temperature was maintained to 33 ± 0.5°C for 2 hours. The results showed that both Drp1 inhibition and therapeutic hypothermia increased 3-day survival time (all P <0.05) and improved neurologic function up to 72 hours post cardiac arrest. In addition, both Drp1 inhibition and therapeutic hypothermia decreased cell injury, apoptosis in hippocampal cornu ammonis 1 region and brain mitochondrial dysfunction including adenosine triphosphate production, reactive oxygen species and mitochondrial membrane potential after cardiac arrest. Moreover, therapeutic hypothermia decreased mitochondrial Drp1 expression and mitochondrial fission after cardiac arrest. In conclusion, inhibition of Drp1 has a similar effect to therapeutic hypothermia on neurologic outcome after resuscitation in this cardiac arrest rat model, and the neuroprotective effects of therapeutic hypothermia are associated with inhibition of mitochondrial fission.


Subject(s)
Dynamins/antagonists & inhibitors , Heart Arrest/therapy , Hypothermia, Induced , Neuroprotective Agents/pharmacology , Quinazolinones/pharmacology , Animals , Disease Models, Animal , Male , Mitochondrial Dynamics/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
11.
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
12.
Int J Biol Sci ; 12(8): 1000-9, 2016.
Article in English | MEDLINE | ID: mdl-27489503

ABSTRACT

Mitochondrial dysfunction contributes to brain injury following global cerebral ischemia after cardiac arrest. Carbon monoxide treatment has shown potent cytoprotective effects in ischemia/reperfusion injury. This study aimed to investigate the effects of carbon monoxide-releasing molecules on brain mitochondrial dysfunction and brain injury following resuscitation after cardiac arrest in rats. A rat model of cardiac arrest was established by asphyxia. The animals were randomly divided into the following 3 groups: cardiac arrest and resuscitation group, cardiac arrest and resuscitation plus carbon monoxide intervention group, and sham control group (no cardiac arrest). After the return of spontaneous circulation, neurologic deficit scores (NDS) and S-100B levels were significantly decreased at 24, 48, and 72 h, but carbon monoxide treatment improved the NDS and S-100B levels at 24 h and the 3-day survival rates of the rats. This treatment also decreased the number of damaged neurons in the hippocampus CA1 area and increased the brain mitochondrial activity. In addition, it increased mitochondrial biogenesis by increasing the expression of biogenesis factors including peroxisome proliferator-activated receptor-γ coactivator-1α, nuclear respiratory factor-1, nuclear respiratory factor-2 and mitochondrial transcription factor A. Thus, this study showed that carbon monoxide treatment alleviated brain injury after cardiac arrest in rats by increased brain mitochondrial biogenesis.


Subject(s)
Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Carbon Monoxide/therapeutic use , Heart Arrest/drug therapy , Heart Arrest/metabolism , Mitochondria/metabolism , Organelle Biogenesis , Animals , Brain/drug effects , Brain/metabolism , Brain Ischemia/etiology , DNA-Binding Proteins/metabolism , GA-Binding Protein Transcription Factor/metabolism , Heart Arrest/complications , Male , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Nuclear Respiratory Factor 1/metabolism , PPAR alpha/metabolism , Rats , Transcription Factors/metabolism
13.
J Cardiovasc Pharmacol Ther ; 20(3): 330-41, 2015 May.
Article in English | MEDLINE | ID: mdl-25420477

ABSTRACT

The objective of this study is to examine whether carbon monoxide-releasing molecules (CORMs) can decrease the generation of excessive reactive oxygen species (ROS) in cardiac mitochondria, thereby protecting against postresuscitation myocardial injury and cardiac mitochondrial dysfunction after resuscitation in a rat model of ventricular fibrillation (VF), and further investigated the underlying mechanism. Rats suffered 8 minutes of untreated VF and resuscitation and were randomized into the control group with vehicle infusion and the CORM group with CO-releasing molecule 2 (CORM2) treatment. Animals in the Sham group were instrumented without induced VF and resuscitation. Effects of CORM2 on cardiac function, myocardial oxidative stress, cardiac mitochondrial function, and mitochondrial ROS generation were assessed. Moreover, to further evaluate the direct effect of CORM2 on cardiac mitochondria isolated from resuscitated rats, we measured mitochondrial function and ROS generation when isolated cardiac mitochondria were directly incubated with different concentrations of (CORM2). Compared with the Sham group, the control and CORM groups demonstrated impaired cardiac function, increased myocardial injury, and aggravated mitochondrial damage. CORM2 improved cardiac performance and attenuated myocardial damage and oxidative stress in resuscitated rats. Additionally, animals with CORM2 treatment showed the decreased generation of cardiac mitochondrial ROS, alleviated mitochondrial injury, and preserved mitochondrial function and complex activities when compared with the control group. In isolated cardiac mitochondria incubated with CORM2, low concentrations of CORM2 (20 µmol/L) mildly uncoupled mitochondrial respiration, leading to reduced mitochondrial ROS production. In contrast, high concentrations of CORM2 (60 µmol/L) resulted in the reverse effect presumably due to its excessive uncoupling action. These findings suggest that CORM2 attenuates oxidative stress of the heart and improves cardiac function after resuscitation. The mechanism was probably that CO, the product of CORM2, reduces the production of cardiac mitochondrial ROS and thereby attenuates mitochondrial injury and dysfunction during the postresuscitation period, due to the transient uncoupling of mitochondrial respiration.


Subject(s)
Carbon Monoxide/pharmacology , Heart Arrest/drug therapy , Mitochondria, Heart/metabolism , Reactive Oxygen Species/metabolism , Animals , Arterial Pressure/drug effects , Heart Arrest/metabolism , Male , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Resuscitation
14.
World J Emerg Med ; 5(2): 116-21, 2014.
Article in English | MEDLINE | ID: mdl-25215160

ABSTRACT

BACKGROUND: Partial pressure of end-tidal carbon dioxide (PETCO2) has been used to monitor the effectiveness of precordial compression (PC) and regarded as a prognostic value of outcomes in cardiopulmonary resuscitation (CPR). This study was to investigate changes of PETCO2 during CPR in rats with ventricular fibrillation (VF) versus asphyxial cardiac arrest. METHODS: Sixty-two male Sprague-Dawley (SD) rats were randomly divided into an asphyxial group (n=32) and a VF group (n=30). PETCO2 was measured during CPR from a 6-minute period of VF or asphyxial cardiac arrest. RESULTS: The initial values of PETCO2 immediately after PC in the VF group were significantly lower than those in the asphyxial group (12.8±4.87 mmHg vs. 49.2±8.13 mmHg, P=0.000). In the VF group, the values of PETCO2 after 6 minutes of PC were significantly higher in rats with return of spontaneous circulation (ROSC), compared with those in rats without ROSC (16.5±3.07 mmHg vs. 13.2±2.62 mmHg, P=0.004). In the asphyxial group, the values of PETCO2 after 2 minutes of PC in rats with ROSC were significantly higher than those in rats without ROSC (20.8±3.24 mmHg vs. 13.9±1.50 mmHg, P=0.000). Receiver operator characteristic (ROC) curves of PETCO2 showed significant sensitivity and specificity for predicting ROSC in VF versus asphyxial cardiac arrest. CONCLUSIONS: The initial values of PETCO2 immediately after CPR may be helpful in differentiating the causes of cardiac arrest. Changes of PETCO2 during CPR can predict outcomes of CPR.

15.
Biomed Res Int ; 2014: 192769, 2014.
Article in English | MEDLINE | ID: mdl-24696844

ABSTRACT

Postcardiac arrest brain injury significantly contributes to mortality and morbidity in patients suffering from cardiac arrest (CA). Evidence that shows that mitochondrial dysfunction appears to be a key factor in tissue damage after ischemia/reperfusion is accumulating. However, limited data are available regarding the cerebral mitochondrial dysfunction during CA and cardiopulmonary resuscitation (CPR) and its relationship to the alterations of high-energy phosphate. Here, we sought to identify alterations of mitochondrial morphology and oxidative phosphorylation function as well as high-energy phosphates during CA and CPR in a rat model of ventricular fibrillation (VF). We found that impairment of mitochondrial respiration and partial depletion of adenosine triphosphate (ATP) and phosphocreatine (PCr) developed in the cerebral cortex and hippocampus following a prolonged cardiac arrest. Optimal CPR might ameliorate the deranged phosphorus metabolism and preserve mitochondrial function. No obvious ultrastructural abnormalities of mitochondria have been found during CA. We conclude that CA causes cerebral mitochondrial dysfunction along with decay of high-energy phosphates, which would be mitigated with CPR. This study may broaden our understanding of the pathogenic processes underlying global cerebral ischemic injury and provide a potential therapeutic strategy that aimed at preserving cerebral mitochondrial function during CA.


Subject(s)
Brain/metabolism , Cardiopulmonary Resuscitation , Mitochondria/metabolism , Oxidative Phosphorylation , Ventricular Fibrillation/metabolism , Animals , Cell Respiration , Disease Models, Animal , Energy Metabolism , Hemodynamics , Hippocampus/metabolism , Male , Mitochondria/ultrastructure , Rats, Sprague-Dawley , Ventricular Fibrillation/pathology , Ventricular Fibrillation/physiopathology
16.
Mol Med Rep ; 9(2): 754-62, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24337106

ABSTRACT

Carbon monoxide (CO) is an endogenous gaseous transmitter that exerts multi-protection in ischemia/reperfusion (I/R) injury, but few experimental studies regarding CO on myocardial I/R-induced apoptosis, as well as its underlying mechanism have been conducted. The present study was designed to investigate whether CO released from CO-releasing molecule-2 (CORM-2) is capable of ameliorating myocardial I/R-induced apoptosis via a mitochondrial apoptotic pathway. Primary cultures of neonatal rat cardiomyocytes were randomly distributed into four groups: Control, I/R (cultured cardiomyocytes were subjected to 2 h simulated ischemia followed by 4 h reperfusion), CORM-2 and inactive CORM-2 (iCORM-2) groups (20 µM CORM-2 and 20 µM iCORM-2 were administered at the beginning of reperfusion following ischemia, respectively). Flow cytometric analysis showed that CORM-2 treatment significantly decreased apoptosis of cardiomyocytes triggered by simulated I/R. CORM-2 partially recovered mitochondrial respiration and ultrastructure alteration, and lowered caspase-3 expression and the release of cytochrome c. Furthermore, CORM-2 partly reduced BAK/BAX expression in mitochondria, as well as the BAX level in the cytoplasm. Cardioprotection is lost when CORM-2 is replaced by iCORM-2. CORM-2 treatment, at the time of reperfusion, was concluded to attenuate myocardial I/R-induced apoptosis. The protection mechanisms may be targeted to the mitochondria and involved in the inhibition of the BAK/BAX­mediated intrinsic pathway.


Subject(s)
Apoptosis/genetics , Carbon Monoxide/metabolism , Myocytes, Cardiac/metabolism , Organometallic Compounds/metabolism , Reperfusion Injury/genetics , Animals , Caspase 3/biosynthesis , Cytochromes c/biosynthesis , Gene Expression Regulation , Humans , Metabolic Networks and Pathways/genetics , Mitochondria/metabolism , Myocytes, Cardiac/pathology , Rats , Reperfusion Injury/pathology , bcl-2 Homologous Antagonist-Killer Protein/biosynthesis , bcl-2-Associated X Protein/biosynthesis
17.
Acta Pharmacol Sin ; 34(6): 784-92, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23474707

ABSTRACT

AIM: To investigate the effects of mesenchymal stem cells (MSCs) transplantation on rat global cerebral ischemia and the underlying mechanisms. METHODS: Adult male SD rats underwent asphxial cardiac arrest to induce global cerebral ischemia, then received intravenous injection of 5×10(6) cultured MSCs of SD rats at 2 h after resuscitation. In another group of cardiac arrest rats, tumor necrosis factor-α-induced protein 6 (TSG-6, 6 µg) was injected into the right lateral ventricle. Functional outcome was assessed at 1, 3, and 7 d after resuscitation. Donor MSCs in the brains were detected at 3 d after resuscitation. The level of serum S-100B and proinflammatory cytokines in cerebral cortex were assayed using ELISA. The expression of TSG-6 and proinflammatory cytokines in cerebral cortex was assayed using RT-PCR. Western blot was performed to determine the levels of TSG-6 and neutrophil elastase in cerebral cortex. RESULTS: MSCs transplantation significantly reduced serum S-100B level, and improved neurological function after global cerebral ischemia compared to the PBS-treated group. The MSCs injected migrated into the ischemic brains, and were observed mainly in the cerebral cortex. Furthermore, MSCs transplantation significantly increased the expression of TSG-6, and reduced the expression of neutrophil elastase and proinflammatory cytokines in the cerebral cortex. Intracerebroventricular injection of TSG-6 reproduced the beneficial effects of MSCs transplantation in rats with global cerebral ischemia. CONCLUSION: MSCs transplantation improves functional recovery and reduces inflammatory responses in rats with global cerebral ischemia, maybe via upregulation of TSG-6 expression.


Subject(s)
Brain Ischemia/therapy , Cell Adhesion Molecules/genetics , Inflammation/therapy , Mesenchymal Stem Cell Transplantation/methods , Animals , Blotting, Western , Brain Ischemia/physiopathology , Cell Adhesion Molecules/administration & dosage , Cell Adhesion Molecules/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Inflammation/pathology , Inflammation Mediators/metabolism , Injections, Intravenous , Leukocyte Elastase/genetics , Leukocyte Elastase/metabolism , Male , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , S100 Calcium Binding Protein beta Subunit/blood , Time Factors , Treatment Outcome , Up-Regulation
18.
Indian J Med Res ; 138(6): 904-11, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24521634

ABSTRACT

BACKGROUND & OBJECTIVES: The gut contains some endogenous and exogenous microorganisms that can become potential pathogens of sepsis under certain circumstances. Therefore, the integrity and normal function of gut barrier is important for preventing the development of sepsis. The present study was designed to assess the effects of ulinastatin, a urinary trypsin inhibitor on gut barrier function and mortality in experimental sepsis. METHODS: Male Sprague-Dawley rats were subjected to ceacal ligation and puncture (CLP) or sham procedure. Rats were then treated with ulinastatin 50,000 U/kg/day or saline. The mortality rate was determined. Histology, apoptosis assays, and PCR were performed using ileum specimens at 3, 6, and 12 h following CLP. Serum levels of tumour necrosis factor α (TNF-α) and interleukin-6 (IL-6) were also measured at 0, 3, 6, and 12 h following CLP. RESULTS: Compared with the saline-treated CLP rats, the ulinastatin CLP rats had significantly increased survival time (P<0.05), lower histopathological scores of intestinal injury (P<0.05), reduced apoptosis detected by terminal deoxynucleotidyl transferase dUTP nick end labelling assay and caspase 3 activity (P<0.01). Moreover, RD-5 mRNA expression was significantly higher in ulinastatin-treated CLP animals than saline controls (P<0.05). These results suggested a preserved integrity and function of the gut barrier. Significantly lower plasma TNFα and IL-6 levels were detected in CLP rats with ulinastatin treatment, which contributed to increased survival time. INTERPRETATION & CONCLUSIONS: Our results suggest that ulinastatin has a therapeutic potential to prevent gut barrier dysfunction in the early stage of sepsis, thereby improving the outcome of sepsis. Further studies need to be done to understand the mechanism of action of ulinastatin.


Subject(s)
Apoptosis/drug effects , Gastrointestinal Tract/microbiology , Glycoproteins/administration & dosage , Sepsis/drug therapy , Animals , Gastrointestinal Tract/pathology , Interleukin-6/blood , Male , Rats, Sprague-Dawley , Sepsis/blood , Sepsis/microbiology , Tumor Necrosis Factor-alpha/blood
19.
Biomed Res Int ; 2013: 564501, 2013.
Article in English | MEDLINE | ID: mdl-24455704

ABSTRACT

Patients with chronic left ventricular (LV) dysfunction are assumed to have a lower chance of successful CPR and lower likelihood of ultimate survival. However, these assumptions have rarely been documented. Therefore, we investigated the outcome of prolonged ventricular fibrillation (VF) and CPR in a rat model of chronic LV dysfunction. Sprague-Dawley rats were randomized to (1) chronic LV dysfunction: animals underwent left coronary artery ligation; and (2) sham control. Echocardiography was used to measure cardiac performance before surgery and 4 weeks after surgery. Four weeks after surgical intervention, 8 min of VF was induced and defibrillation was delivered after 8 min of CPR. LV dilation and low ejection fraction were observed 4 weeks after coronary ligation. With optimal chest compressions, coronary perfusion pressure values during CPR were well maintained and indistinguishable between groups. There were no differences in resuscitability and numbers of shock required for successful resuscitation between groups. Despite the significantly decreased cardiac index in LV dysfunction animals before induction of VF, no differences in cardiac index were observed between groups following resuscitation, which was associated with the insignificant difference in postresuscitation survival. In conclusion, the outcomes of CPR were not compromised by the preexisting chronic LV dysfunction.


Subject(s)
Cardiopulmonary Resuscitation/methods , Coronary Vessels/physiopathology , Heart/physiopathology , Ventricular Dysfunction, Left/physiopathology , Ventricular Fibrillation/physiopathology , Animals , Echocardiography , Ischemia/physiopathology , Ligation , Rats , Treatment Outcome , Ventricular Dysfunction, Left/therapy , Ventricular Fibrillation/therapy
20.
Zhonghua Yi Xue Za Zhi ; 92(15): 1023-7, 2012 Apr 17.
Article in Chinese | MEDLINE | ID: mdl-22781641

ABSTRACT

OBJECTIVE: To explore the effects and mechanisms of heme oxygenase-1 on rats with postresuscitation myocardial dysfunction. METHODS: Male Sprague-Dawley rats were asphyxiated for 9 minutes and resuscitated. They were randomly divided into 4 groups: sham-operated, cardiopulmonary resuscitation (CPR), hemin and hemin + ZnPP (zinc protoporphyrin IX). Resuscitated groups had 2 observation points: 6 and 24 hours post-CPR (n = 8 for each time point). And the sham-operated group of 12 rats were divided in two observation points, according to 6 or 24 hours post-operation (n = 6 each). Hemodynamic was observed. The expression of heme oxygenase-1 (HO-1) in cardiac tissue was detected by Western blot. And the activity of cardiac homogenate superoxide dismutase (SOD) was determined by xanthine oxidase method and the level of malondialdehyde (MDA) measured by the thiobarbituric acid method. Nitrotyrosine protein expression in cardiac tissue was analyzed by immunohistochemistry. RESULTS: (1) The mean blood pressure (MAP) significantly decreased in resuscitated groups after resuscitation (all P < 0.05). No difference existed between the subgroups. The scores of dP/dt40 and -dP/dt significantly decreased in CPR and hemin + ZnPP groups after resuscitation (all P < 0.05). But dP/dt40 in hemin group did not differ significantly after resuscitation and -dP/dt decreased only 0.5 hour and 1 hour post-resuscitation (3341.60 ± 524.85 and 3711.40 ± 502.39 vs 4284.20 ± 800.87, all P < 0.05). The scores of dP/dt40 and -dP/dt in hemin group at all time points post-resuscitation were significantly higher than those in CPR and hemin + ZnPP groups (all P < 0.05). (2) Compared with the sham-operated group, the HO-1 expression, MDA level and nitrotyrosine protein expression significantly increased while the activities of SOD decreased after resuscitation in the CPR, hemin and hemin + ZnPP groups (all P < 0.05). Compared with the CPR and hemin + ZnPP groups, the expression of HO-1 and the activity of SOD increased, while MDA level and nitrotyrosine protein expression were decreased in group hemin (all P < 0.05). No difference existed in the above indices between the CPR and hemin + ZnPP groups. CONCLUSION: HO-1 can reduce myocardial oxidative stress injury after cardiopulmonary resuscitation and effectively improve post-resuscitation myocardial function in rats.


Subject(s)
Asphyxia/physiopathology , Heme Oxygenase (Decyclizing)/metabolism , Myocardium/metabolism , Oxidative Stress , Animals , Asphyxia/metabolism , Asphyxia/therapy , Cardiopulmonary Resuscitation , Male , Malondialdehyde/metabolism , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL