Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters

Affiliation country
Publication year range
1.
Rev Cardiovasc Med ; 25(3): 105, 2024 Mar.
Article in English | MEDLINE | ID: mdl-39076959

ABSTRACT

Microvascular obstruction (MVO) of coronary arteries promotes an increase in mortality and major adverse cardiac events in patients with acute myocardial infarction (AMI) and percutaneous coronary intervention (PCI). Intramyocardial hemorrhage (IMH) is observed in 41-50% of patients with ST-segment elevation myocardial infarction and PCI. The occurrence of IMH is accompanied by inflammation. There is evidence that microthrombi are not involved in the development of MVO. The appearance of MVO is associated with infarct size, the duration of ischemia of the heart, and myocardial edema. However, there is no conclusive evidence that myocardial edema plays an important role in the development of MVO. There is evidence that platelets, inflammation, Ca 2 + overload, neuropeptide Y, and endothelin-1 could be involved in the pathogenesis of MVO. The role of endothelial cell damage in MVO formation remains unclear in patients with AMI and PCI. It is unclear whether nitric oxide production is reduced in patients with MVO. Only indirect evidence on the involvement of inflammation in the development of MVO has been obtained. The role of reactive oxygen species (ROS) in the pathogenesis of MVO is not studied. The role of necroptosis and pyroptosis in the pathogenesis of MVO in patients with AMI and PCI is also not studied. The significance of the balance of thromboxane A2, vasopressin, angiotensin II, and prostacyclin in the formation of MVO is currently unknown. Conclusive evidence regarding the role of coronary artery spasm in the development of MVhasn't been established. Correlation analysis of the neuropeptide Y, endothelin-1 levels and the MVO size in patients with AMI and PCI has not previously been performed. It is unclear whether epinephrine aggravates reperfusion necrosis of cardiomyocytes. Dual antiplatelet therapy improves the efficacy of PCI in prevention of MVO. It is unknown whether epinephrine or L-type Ca 2 + channel blockers result in the long-term improvement of coronary blood flow in patients with MVO.

2.
Med Res Rev ; 36(5): 871-923, 2016 09.
Article in English | MEDLINE | ID: mdl-27197922

ABSTRACT

It has now been demonstrated that the µ, δ1 , δ2 , and κ1 opioid receptor (OR) agonists represent the most promising group of opioids for the creation of drugs enhancing cardiac tolerance to the detrimental effects of ischemia/reperfusion (I/R). Opioids are able to prevent necrosis and apoptosis of cardiomyocytes during I/R and improve cardiac contractility in the reperfusion period. The OR agonists exert an infarct-reducing effect with prophylactic administration and prevent reperfusion-induced cardiomyocyte death when ischemic injury of heart has already occurred; that is, opioids can mimic preconditioning and postconditioning phenomena. Furthermore, opioids are also effective in preventing ischemia-induced arrhythmias.


Subject(s)
Analgesics, Opioid/pharmacology , Anti-Arrhythmia Agents/pharmacology , Cardiotonic Agents/pharmacology , Receptors, Opioid/agonists , Analgesics, Opioid/chemistry , Animals , Anti-Arrhythmia Agents/chemistry , Cardiotonic Agents/chemistry , Drug Discovery , Humans
3.
Cytokine ; 71(1): 60-5, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25226445

ABSTRACT

This study was designed to investigate whether lacidipine elicited a protective role on cardiomyocyte against apoptosis induced by TNF-α. Neonatal rat cardiomyocytes were randomly assigned into different groups. TUNEL staining was utilized to detect apoptosis, and caspase-3 and caspse-12 were determined. To explore the underlying mechanism, Z-ATAD-FMK (a selective caspase-12 inhibitor) was used to identify the key molecule involved. TNF-α increased caspase-3 expression, which was mediated by increased caspase-12 expression. In the meantime, apoptosis was significantly induced by TNF-α. Lacidipine lowered caspase-12 and caspase-3 expression, and cardiomyocyte apoptosis induced by TNF-α. The results suggest that lacidipine attenuates TNF-α -induced apoptosis via inhibition of caspase-12 and caspase-3 successively.


Subject(s)
Apoptosis/drug effects , Calcium Channel Blockers/pharmacology , Dihydropyridines/pharmacology , Myocytes, Cardiac/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Caspase 12/genetics , Caspase 12/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Cells, Cultured , In Situ Nick-End Labeling , Male , Myocytes, Cardiac/physiology , Random Allocation , Rats, Sprague-Dawley
4.
Fundam Clin Pharmacol ; 38(3): 489-501, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38311344

ABSTRACT

BACKGROUND: The high mortality rate of patients with acute myocardial infarction (AMI) remains the most pressing issue of modern cardiology. Over the past 10 years, there has been no significant reduction in mortality among patients with AMI. It is quite obvious that there is an urgent need to develop fundamentally new drugs for the treatment of AMI. Angiotensin 1-7 has some promise in this regard. OBJECTIVE: The objective of this article is analysis of published data on the cardioprotective properties of angiotensin 1-7. METHODS: PubMed, Scopus, Science Direct, and Google Scholar were used to search articles for this study. RESULTS: Angiotensin 1-7 increases cardiac tolerance to ischemia/reperfusion and mitigates adverse remodeling of the heart. Angiotensin 1-7 can prevent not only ischemic but also reperfusion cardiac injury. The activation of the Mas receptor plays a key role in these effects of angiotensin 1-7. Angiotensin 1-7 alleviates Ca2+ overload of cardiomyocytes and reactive oxygen species production in ischemia/reperfusion (I/R) of the myocardium. It is possible that both effects are involved in angiotensin 1-7-triggered cardiac tolerance to I/R. Furthermore, angiotensin 1-7 inhibits apoptosis of cardiomyocytes and stimulates autophagy of cells. There is also indirect evidence suggesting that angiotensin 1-7 inhibits ferroptosis in cardiomyocytes. Moreover, angiotensin 1-7 possesses anti-inflammatory properties, possibly achieved through NF-kB activity inhibition. Phosphoinositide 3-kinase, Akt, and NO synthase are involved in the infarct-reducing effect of angiotensin 1-7. However, the specific end-effector of the cardioprotective impact of angiotensin 1-7 remains unknown. CONCLUSION: The molecular nature of the end-effector of the infarct-limiting effect of angiotensin 1-7 has not been elucidated. Perhaps, this end-effector is the sarcolemmal KATP channel or the mitochondrial KATP channel.


Subject(s)
Angiotensin I , Myocardial Reperfusion Injury , Peptide Fragments , Signal Transduction , Angiotensin I/pharmacology , Peptide Fragments/pharmacology , Humans , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/physiopathology , Animals , Signal Transduction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Ventricular Remodeling/drug effects , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Apoptosis/drug effects
5.
Am J Ther ; 20(5): 493-501, 2013.
Article in English | MEDLINE | ID: mdl-23011166

ABSTRACT

κ-opioid receptor (κ-OR) activation with U50,488H, a selective κ-OR agonist, has been previously demonstrated to prevent against cardiac arrhythmias via stabilizing the synthesis and degradation of an integral membrane protein, Cx43, in gap junctions. However, the exact prevention mechanism remains unclear. The present study tested the hypothesis that the kappa OR agonist U50,488H mediates the prevention of arrhythmia through the regulation of intracellular calcium leading to the preservation of Cx43 protein. By performing electrocardiogram monitoring and immunoblotting in isolated Langendorff-perfused rat hearts, high concentrations of calcium-perfused rat hearts exhibited increased cardiac arrhythmias. Diminished expression of Cx43 protein was observed. The utilization of a whole-cell patch clamp technique revealed that U50,488H inhibited L-type calcium current in single ventricular myocytes in a dose-dependent manner. These effects were blocked by nor-binaltorphimine, potent and selective κ-OR antagonists. Administration of U50,488H before myocardial ischemia resulted in an attenuated of total arrhythmia scores. The attenuation effect was blocked by nor-binaltorphimine. The attenuation effect was antagonized both by Bay K8644, a L-type calcium channel agonist, and also by the Cx43 uncoupler heptanol. Finally, immunoblotting data demonstrated that the preservation of Cx43 protein conferred by U50,488H was reversed in the presence of Bay K8644. In summary, the present study demonstrates κ-OR activation with U50,488H may confer antiarrhythmic effects via modulation of the calcium-Cx43 pathway.


Subject(s)
3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Antihypertensive Agents/pharmacology , Arrhythmias, Cardiac/prevention & control , Connexin 43/metabolism , Receptors, Opioid, kappa/agonists , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Calcium/metabolism , Calcium Channel Agonists/pharmacology , Dose-Response Relationship, Drug , Male , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/antagonists & inhibitors
6.
Biochim Biophys Acta Gen Subj ; 1867(2): 130281, 2023 02.
Article in English | MEDLINE | ID: mdl-36410609

ABSTRACT

BACKGROUND: Uncorrected obesity facilitates premature aging and cardiovascular anomalies. This study examined the interaction between obesity and aging on cardiac remodeling and contractile function. METHODS: Cardiac echocardiographic geometry, function, morphology, intracellular Ca2+ handling, oxidative stress (DHE fluorescence), STAT3 and stress signaling were evaluated in young (3-mo) and old (12- and 18-mo) lean and leptin deficient ob/ob obese mice. Cardiomyocytes from young and old lean and ob/ob mice were treated with leptin (1 nM) for 4 h in vitro prior to assessment of mechanical and biochemical properties. High fat diet (45% calorie from fat) and the leptin receptor mutant db/db obese mice at young and old age were evaluated for comparison. RESULTS: Our results displayed reduced survival in ob/ob mice. Obesity but less likely older age dampened echocardiographic, geometric, cardiomyocyte function and intracellular Ca2+ properties, elevated O2- and p47phox NADPH oxidase levels with a more pronounced geometric change at older age. Immunoblot analysis revealed elevated p47phox NADPH oxidase and dampened phosphorylation of STAT3, with a more pronounced response in old ob/ob mice, the effects were restored by leptin. Obesity and aging inhibited phosphorylation of Akt, eNOS, AMPK, and p38 while promoting phosphorylation of JNK and IκB. Leptin reconciled cardiomyocyte dysfunction, O2- yield, p47phox upregulation, STAT3 dephosphorylation and stress signaling in ob/ob mice although its action on stress signaling cascades were lost at old age. High fat diet-induced and db/db obesity displayed aging-associated cardiomyocyte anomalies reminiscent of ob/ob model albeit lost leptin response. CONCLUSIONS: Our data suggest disparate age-associated obesity response in cardiac remodeling and contractile dysfunction due to phosphorylation of Akt, eNOS and stress signaling-related oxidative stress.


Subject(s)
Aging , Leptin , Myocardium , Obesity , Animals , Mice , Leptin/physiology , Mice, Obese , NADPH Oxidases , Proto-Oncogene Proteins c-akt , Ventricular Remodeling , Myocardium/pathology , Oxidative Stress , Stress, Physiological
7.
Fundam Clin Pharmacol ; 37(6): 1020-1049, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37218378

ABSTRACT

BACKGROUND: The use of percutaneous coronary intervention (PCI) in patients with ST-segment elevation myocardial infarction (STEMI) is associated with a mortality rate of 5%-7%. It is clear that there is an urgent need to develop new drugs that can effectively prevent cardiac reperfusion injury. ATP-sensitive K+ (KATP ) channel openers (KCOs) can be classified as such drugs. RESULTS: KCOs prevent irreversible ischemia and reperfusion injury of the heart. KATP channel opening promotes inhibition of apoptosis, necroptosis, pyroptosis, and stimulation of autophagy. KCOs prevent the development of cardiac adverse remodeling and improve cardiac contractility in reperfusion. KCOs exhibit antiarrhythmic properties and prevent the appearance of the no-reflow phenomenon in animals with coronary artery occlusion and reperfusion. Diabetes mellitus and a cholesterol-enriched diet abolish the cardioprotective effect of KCOs. Nicorandil, a KCO, attenuates major adverse cardiovascular event and the no-reflow phenomenon, reduces infarct size, and decreases the incidence of ventricular arrhythmias in patients with acute myocardial infarction. CONCLUSION: The cardioprotective effect of KCOs is mediated by the opening of mitochondrial KATP (mitoKATP ) and sarcolemmal KATP (sarcKATP ) channels, triggered free radicals' production, and kinase activation.


Subject(s)
Myocardial Reperfusion Injury , No-Reflow Phenomenon , Percutaneous Coronary Intervention , Humans , Animals , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/prevention & control , Apoptosis , Reperfusion , Adenosine Triphosphate , KATP Channels
8.
J Biomed Res ; 36(6): 375-389, 2022 Aug 10.
Article in English | MEDLINE | ID: mdl-36320147

ABSTRACT

Acute myocardial infarction (AMI) is one of the main reasons of cardiovascular disease-related death. The introduction of percutaneous coronary intervention to clinical practice dramatically decreased the mortality rate in AMI. Adverse cardiac remodeling is a serious problem in cardiology. An increase in the effectiveness of AMI treatment and prevention of adverse cardiac remodeling is difficult to achieve without understanding the mechanisms of reperfusion cardiac injury and cardiac remodeling. Inhibition of pyroptosis prevents the development of postinfarction and pressure overload-induced cardiac remodeling, and mitigates cardiomyopathy induced by diabetes and metabolic syndrome. Therefore, it is reasonable to hypothesize that the pyroptosis inhibitors may find a role in clinical practice for treatment of AMI and prevention of cardiac remodeling, diabetes and metabolic syndrome-triggered cardiomyopathy. It was demonstrated that pyroptosis interacts closely with apoptosis and autophagy. Pyroptosis could be inhibited by nucleotide-binding oligomerization domain-like receptor with a pyrin domain 3 inhibitors, caspase-1 inhibitors, microRNA, angiotensin-converting enzyme inhibitors, angiotensin Ⅱ receptor blockers, and traditional Chinese herbal medicines.

9.
Curr Cardiol Rev ; 18(5): 63-79, 2022.
Article in English | MEDLINE | ID: mdl-35422224

ABSTRACT

It has been documented that Ca2+ overload and increased production of reactive oxygen species play a significant role in reperfusion injury (RI) of cardiomyocytes. Ischemia/reperfusion induces cell death as a result of necrosis, necroptosis, apoptosis, and possibly autophagy, pyroptosis and ferroptosis. It has also been demonstrated that the NLRP3 inflammasome is involved in RI of the heart. An increase in adrenergic system activity during the restoration of coronary perfusion negatively affected cardiac resistance to RI. Toll-like receptors are involved in RI of the heart. Angiotensin II and endothelin-1 aggravated ischemic/reperfusion injury of the heart. Activation of neutrophils, monocytes, CD4+ T-cells and platelets contributes to cardiac ischemia/reperfusion injury. Our review outlines the role of these factors in reperfusion cardiac injury.


Subject(s)
Inflammasomes , Reperfusion Injury , Adrenergic Agents/metabolism , Angiotensin II/metabolism , Endothelin-1/metabolism , Humans , Inflammasomes/metabolism , Ischemia/metabolism , Myocytes, Cardiac/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reactive Oxygen Species/metabolism , Reperfusion
10.
J Cardiovasc Pharmacol ; 58(2): 207-15, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21654505

ABSTRACT

The aim of this study was to investigate the underlying mechanism that dynorphin, an endogenous kappa opioid receptor (κ-OR) agonist, triggers antiapoptotic effect of postconditioning (Postcon). In addition to vehicle treatment, Sprague Dawley rats (n = 6) underwent a 30-minute left anterior descending occlusion followed by 2 hours of reperfusion with or without a Postcon stimulus. The selective κ-OR antagonist nor-binaltorphimine (Nor-BNI) was administered intravenously 5 minutes before reperfusion. Infarct size was determined by using 2,3,5-triphenyltetrazolium chloride staining. Blood plasma concentrations of creatine kinase (CK) and lactate dehydrogenase (LDH) and myocardial caspase-3 activity were analyzed spectrophotometrically. Myocardial apoptosis was analyzed by the detection of terminal deoxynucleotidyl transferase (TdT)-mediated deoxyuridine triphosphate (dUTP) nick-end labeling. Immunoreactive dynorphin in blood serum and myocardium was measured by means of an antigen-competitive enzyme-linked immunosorbent assay. Infarction size, caspase-3 activity, apoptotic index, and CK and LDH levels were significantly higher in the ischemic/reperfusion group than in the vehicle group (P < 0.01). Postcon significantly reduced infarction size, caspase-3 activity, apoptotic index, CK and LDH levels (P < 0.01 vs. ischemic/reperfusion). Dynorphin content significantly increased after Postcon (P < 0.01). All the effects described above were abolished by Nor-BNI, with the exception of dynorphin content. We found that cardiac protection and antiapoptotic effect of Postcon is mediated by the activation of κ-OR. Effect of Postcon is mediated, at least partially, by enhanced dynorphin expression.


Subject(s)
Apoptosis , Dynorphins/metabolism , Ischemic Postconditioning , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Receptors, Opioid, kappa/agonists , Animals , Disease Models, Animal , Dynorphins/blood , Hemodynamics , In Situ Nick-End Labeling , Male , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Rats , Rats, Sprague-Dawley
11.
Crit Care Med ; 38(12): 2365-76, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20890194

ABSTRACT

OBJECTIVE: Acute myocardial ischemia induces electrical and chemical uncoupling of gap junctions, which contributes to conduction abnormalities and re-entrant arrhythmias. We tested the hypothesis that structure and function of Connexin43 may vibrate during acute myocardial ischemia and reperfusion and κ-opioid receptor stimulation may stabilize the alteration of Connexin43. DESIGN: An animal intervention study was conducted with comparison to a control group. SETTING: University preclinical research laboratory. SUBJECTS: Age-, weight-, and sex-matched Sprague-Dawley rats. INTERVENTIONS: Adult rat hearts were subjected to ischemia or ischemia/reperfusion, which was induced by temporary occlusion of the left main coronary artery. U50488H was given 10 mins before tissue specimens were taken or before ischemia (1.5 mg/kg, intravenous) and nor-BNI was given 15 mins before tissue specimens were taken or before ischemia (2 mg/kg, intravenous). Tissue samples came from left ventricular myocardium of the rat hearts. MEASUREMENTS AND MAIN RESULTS: Electrocardiogram, immunohistochemistry, immunoblotting, and reverse transcription-polymerase chain reaction were used to measure changes of arrhythmias, protein, and gene expression of Connexin43, respectively. κ-opioid receptor activation with U50 decreased arrhythmia in a model of myocardial ischemia and reperfusion. In normal hearts, immunohistochemical data showed reduced amount and lateralization of Connexin43 induced by κ-opioid receptor activation, whereas immunoblotting data demonstrated no significant changes between control and U50 group. During ischemia, however, Connexin43 protein underwent dephosphorylation and degradation, and Connexin43 mRNA was upregulated. These alterations were significantly attenuated on κ-opioid receptor stimulation. During ischemia and reperfusion, Connexin43 protein underwent dephosphorylation and degradation and recovered slowly during reperfusion. Activation of κ-opioid receptor accelerated recovery of phosphorylated and total Connexin43. CONCLUSIONS: In normal rat hearts, Connexin43 translocates from intercellular junctions to intracellular locations on κ-opioid receptor activation. In rat hearts experiencing acute myocardial ischemia and reperfusion, protein and gene expression of Connexin43 undergo vibration. This phenomenon is stabilized when κ-opioid receptor is activated and by the fact that κ-opioid receptor produces antiarrhythmic effects.


Subject(s)
3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Arrhythmias, Cardiac/drug therapy , Connexin 43/metabolism , Myocardial Ischemia/drug therapy , Myocardial Reperfusion Injury/drug therapy , Receptors, Opioid, kappa/metabolism , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/metabolism , Animals , Arrhythmias, Cardiac/physiopathology , Blotting, Western , Connexin 43/drug effects , Disease Models, Animal , Female , Gap Junctions/drug effects , Immunohistochemistry , Male , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Random Allocation , Rats , Rats, Sprague-Dawley , Reference Values , Reverse Transcriptase Polymerase Chain Reaction
12.
Theranostics ; 10(5): 2422-2435, 2020.
Article in English | MEDLINE | ID: mdl-32104514

ABSTRACT

Macrophages are essential for wound repair after myocardial infarction (MI). CD226, a member of immunoglobulin superfamily, is expressed on inflammatory monocytes, however, the role of CD226 in infarct healing and the effect of CD226 on macrophage remain unknown. Methods: Wild type and CD226 knockout (CD226 KO) mice were subjected to permanent coronary ligation. CD226 expression, cardiac function and ventricular remodeling were evaluated. Profile of macrophages, myofibroblasts, angiogenesis and monocytes mobilization were determined. Results: CD226 expression increased in the infarcted heart, with a peak on day 7 after MI. CD226 KO attenuated infarct expansion and improved infarct healing after MI. CD226 deletion resulted in increased F4/80+ CD206+ M2 macrophages and diminished Mac-3+ iNOS+ M1 macrophages accumulation in the infarcted heart, as well as enrichment of α-smooth muscle actin positive myofibroblasts and Ki67+ CD31+ endothelial cells, leading to increased reparative collagen deposition and angiogenesis. Furthermore, CD226 deletion restrained inflammatory monocytes mobilization, as revealed by enhanced retention of Ly6Chi monocytes in the spleen associated with a decrease of Ly6Chi monocytes in the peripheral blood, whereas local proliferation of macrophage in the ischemic heart was not affected by CD226 deficiency. In vitro studies using bone marrow-derived macrophages showed that CD226 deletion potentiated M2 polarization and suppressed M1 polarization. Conclusion: CD226 expression is dramatically increased in the infarcted heart, and CD226 deletion improves post-infarction healing and cardiac function by favoring macrophage polarization towards reparative phenotype. Thus, inhibition of CD226 may represent a novel therapeutic approach to improve wound healing and cardiac function after MI.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/metabolism , Macrophages/metabolism , Myocardial Infarction/metabolism , Ventricular Remodeling , Animals , Antigens, Differentiation, T-Lymphocyte/genetics , Endothelial Cells/metabolism , Macrophage Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Phenotype , Wound Healing
13.
Cytokine ; 44(1): 96-100, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18783962

ABSTRACT

To determine whether insulin administration modulates the systemic inflammatory response in infants undergoing cardiac surgery with cardiopulmonary bypass, 60 infants undergoing cardiopulmonary bypass were randomly assigned into a routine therapy group or to an intensive insulin therapy group with 30 infants in each group. Plasma IL-1beta, IL-6, IL-10, and TNF-alpha levels were determined before anesthesia, at the initiation of cardiopulmonary bypass, and at 0, 6, 12, 24, and 48 h after cardiopulmonary bypass. Nuclear factor-kappaBp65 expression and IkappaB expression in peripheral blood mononuclear cells were also measured by Western blot analysis. TNF-alpha, IL-1beta, IL-6, and IL-10 levels were all elevated after the initiation of cardiopulmonary bypass. However, TNF-alpha, IL-1beta, and IL-6 levels were significantly attenuated in the intensive insulin therapy group compared to those in the routine therapy group after initiation of cardiopulmonary bypass (p<0.05 or <0.01). Meanwhile, plasma IL-10 levels were significantly higher in the intensive insulin therapy group than in the routine therapy group after initiation of cardiopulmonary bypass (p<0.05 or <0.01). Accordingly, Nuclear factor-kappaBp65 expression and IkappaB expression were significantly increased after initiation of cardiopulmonary bypass in both groups (p<0.05 or <0.01). The expression of Nuclear factor-kappaBp65, which induces the transcription of pro-inflammatory cytokines was significantly attenuated in the intensive insulin therapy group (p<0.05 or <0.01). Meanwhile, the expression of IkappaB, an inhibitor of NF-kappaB, was significantly higher in the intensive insulin therapy group (p<0.05 or <0.01). These results suggested that intensive insulin therapy may attenuate the systemic inflammatory response in infants undergoing cardiopulmonary bypass.


Subject(s)
Cardiopulmonary Bypass , Inflammation Mediators/blood , Insulin/therapeutic use , Blood Glucose/metabolism , Cardiopulmonary Bypass/adverse effects , Female , Heart Defects, Congenital/surgery , Humans , I-kappa B Proteins/blood , Infant , Inflammation/drug therapy , Insulin/blood , Interleukin-10/blood , Interleukin-1beta/blood , Interleukin-6/blood , Intraoperative Period , Male , Transcription Factor RelA/blood , Tumor Necrosis Factor-alpha/blood
14.
J Appl Physiol (1985) ; 105(2): 569-74, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18511523

ABSTRACT

The modulation of beta-adrenoceptor signaling in the hearts of hindlimb unweighting (HU) simulated weightlessness rats has not been reported. In the present study, we adopted the rat tail suspension for 4 wk to simulate weightlessness; then the effects of simulated microgravity on beta-adrenoceptor signaling were studied. Mean arterial blood pressure (ABP), left ventricular pressure (LVP), systolic function (+dP/dtmax), and diastolic function (-dP/dtmax) were monitored in the course of the in vivo experiment. Single rat ventricular myocyte was obtained by the enzymatic dissociation method. Hemodynamics, myocyte contraction, and cAMP production in response to beta-adrenoceptor stimulation with isoproterenol or adenylyl cyclase stimulation with forskolin were measured, and Gs protein was also determined. Compared with the control group, no significant changes were found in heart weight, body weight and ABP, while LVP and +/-dP/dtmax were significantly reduced. The ABP decrease, LVP increase, and +/-dP/dtmax in response to isoproterenol administration were significantly attenuated in the HU group. The effects of isoproterenol on electrically induced single-cell contraction and cAMP production in myocytes of ventricles in the HU rats were significantly attenuated. The biologically active isoform, Gsalpha (45 kDa) in the heart, was unchanged. Both the increased electrically induced contraction and cAMP production in response to forskolin were also significantly attenuated in the simulated weightlessness rats. Above results indicated that impaired function of adenylyl cyclase causes beta-adrenoceptor desensitization, which may be partly responsible for the depression of cardiac function.


Subject(s)
Heart/physiology , Receptors, Adrenergic, beta/physiology , Signal Transduction/physiology , Weightlessness Simulation , Adenylyl Cyclases/metabolism , Adrenergic beta-Agonists/pharmacology , Anesthesia , Animals , Body Weight/physiology , Colforsin/pharmacology , Cyclic AMP/metabolism , Electric Stimulation , Hindlimb Suspension/physiology , Isoproterenol/pharmacology , Male , Muscle Contraction/physiology , Myocardial Contraction/physiology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Organ Size/physiology , Rats , Rats, Sprague-Dawley
15.
Arch Med Res ; 39(1): 45-51, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18067995

ABSTRACT

BACKGROUND: Multiple sclerosis (MS) is a chronic disease characterized by loss of myelin. However, data indicate that autoimmune cells could directly impair neuronal cell bodies and myelin sheath is lacking. The aim of the present study was to determine morphological evidence of the direct impairment of neurons by autoreactive lymphocytes and to further identify the subtypes of these lymphocytes. METHODS: Lymphocytes activated by myelin basic protein (MBP) 83-99 and neurons of human brain were co-cultured for 24 h. RESULTS: Observations through scanning electron microscope showed that MBP-specific lymphocytes (CD4+, CD8+ cells, and NK cells) aggregated in the vicinity of the neuronal cell bodies and the myelin sheaths and attacked them directly, resulting in the degeneration of both neurons. CONCLUSIONS: Our studies provide morphological evidences of the direct impairment of neuronal cell bodies and myelin sheaths by MBP-specific lymphocytes. Our studies also suggest that MBP-specific CD4+, CD8+, and NK cells might be involved in this process. These processes may play a role in the direct impairment of neurons and myelin sheaths in early stages of MS and provide evidences for the application of immunosuppressant therapy of MS.


Subject(s)
Killer Cells, Natural/immunology , Multiple Sclerosis/immunology , Myelin Basic Protein/immunology , Myelin Sheath/immunology , Neurons/immunology , Adult , CD4 Antigens/analysis , CD8 Antigens/analysis , Coculture Techniques , Female , Humans , Killer Cells, Natural/classification , Killer Cells, Natural/ultrastructure , Male , Middle Aged , Multiple Sclerosis/pathology , Myelin Basic Protein/analysis , Myelin Sheath/ultrastructure , Neurons/ultrastructure , Peptide Fragments/immunology
16.
Am J Chin Med ; 36(3): 541-54, 2008.
Article in English | MEDLINE | ID: mdl-18543387

ABSTRACT

Angelica and ChuanXiong are used to cure ischemic heart disease in China. Previous studies found that these two herbs could increase myocardial blood flow, oxygen-supply and keep myocardial oxygen balance, etc. However, the mechanisms of angiogenic effects of these two herbs are not well-known. The purpose of this study was to assess the effects of Angelica and ChuanXiong on vascular endothelial growth factor (VEGF) expression in rat myocardial infarction, on endothelial cell proliferation and quantity of vessels on chick embryo chorioallantoic membrane (CAM). In this study, rats were divided randomly into either pre-treatment or acute-treatment group and sacrificed at the end of the treatments. VEGF expression using Western blot analysis was significantly increased in the groups pre-treated with ChuanXiong and Angelica when compared to the control group (p < 0.05). There was significant increase in VEGF expression in the rats treated acutely with Angelica (p < 0.05). In the contrary, the rats treated with ChuanXiong showed a decrease in VEGF expression when compared to the acute-treatment control group (p < 0.05). Similar results were observed in immunohistochemistry of VEGF expression in the myocardia. Our study also demonstrated that these two herbs significantly enhanced endothelial cell proliferation (p < 0.05) and revascularity in CAM (p < 0.05). The data showed that Angelica and ChuanXiong could affect VEGF expression in rat myocardial infarction, promote endothelial cell proliferation and stimulate quantity of vessels on CAM model. The results suggest that Angelica and ChuanXiong have angiogenic effects, and may provide some mechanisms for the treatment of myocardial infarction and peripheral ischemia.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Neovascularization, Physiologic/drug effects , Angelica sinensis , Animals , Cell Proliferation/drug effects , Chick Embryo , Chorioallantoic Membrane/blood supply , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/drug effects , Humans , Ligusticum , Male , Myocardial Infarction/metabolism , Neovascularization, Physiologic/physiology , Rats , Rats, Sprague-Dawley , Umbilical Veins/cytology , Umbilical Veins/drug effects , Vascular Endothelial Growth Factor A/metabolism
17.
Eur J Pharmacol ; 572(1): 1-11, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17822695

ABSTRACT

The pathogenesis of myocardial stunning caused by brief ischemia and reperfusion remains unclear. The aim of the present study was to investigate the underlying mechanism of myocardial stunning. An isolated cell model of myocardial stunning was firstly established in isolated rat ventricular myocytes exposed to 8 min of simulated ischemia and 30 min of reperfusion, the cardiomyocyte contractile function was used to evaluate myocardial stunning. A diastolic Ca(2+) overload without significant changes in systolic Ca(2+) and the amplitude of Ca(2+) transient during the first 10 min of reperfusion played an important role in the occurrence of myocardial stunning. Decreasing Ca(2+) entry into myocardial cells with low Ca(2+) reperfusion was a very efficient way to prevent myocardial stunning. Diastolic Ca(2+) overload was closely related to the reverse mode of Na(+)/Ca(2+) exchanger (NCX) rather than L-type Ca(2+) channel. The activity of the reverse mode of NCX was found significantly higher at the initial time of reperfusion, and KB-R7943, a selective inhibitor of the reverse mode of NCX, administered at first 10 min of reperfusion rather than at the time of ischemia significantly attenuated myocardial stunning. In addition, NCX inhibition also attenuated the Ca(2+) oscillation and cardiac dysfunction when field stimulus was stopped at first 10 min of reperfusion. These data suggest that one of the important mechanisms of triggering myocardial stunning is diastolic Ca(2+) overload caused by activation of the reverse mode of NCX of cardiomyocytes during the initial period of reperfusion following brief ischemia.


Subject(s)
Calcium/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardial Stunning/physiopathology , Myocytes, Cardiac/metabolism , Sodium-Calcium Exchanger/metabolism , Animals , Calcium Channels, L-Type/physiology , Diastole , In Vitro Techniques , Male , Myocardial Contraction , Myocardial Reperfusion Injury/metabolism , Myocardial Stunning/metabolism , Myocytes, Cardiac/drug effects , Rats , Rats, Sprague-Dawley , Sodium-Calcium Exchanger/antagonists & inhibitors , Thiourea/administration & dosage , Thiourea/analogs & derivatives , Thiourea/pharmacology
18.
Arch Med Res ; 38(7): 723-9, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17845890

ABSTRACT

BACKGROUND: It remains unclear whether the activation of kappa-opioid receptors has strong hypotensive effects under hypertensive condition, and the underlying mechanisms have not yet been investigated. Therefore, the present study is designed to use spontaneously hypertensive rats (SHR) to investigate the effects of a kappa-opioid receptor agonist on the regulation of urinary formation in hypertensive conditions and to identify its underlying mechanism. METHODS: The hemodynamics, urine flow rate, vasodilatation of isolated renal artery, and plasma hormones were determined by physiological in vivo experimental technique, isolated artery perfusion technique and radioimmunoassay. RESULTS: Intravenous administration of U50, 448H significantly decreased mean arterial blood pressure in both Wistar-Kyoto (WKY) rats and SHR. However, the blood pressure vasodepressor effect of U50, 448H was much more profound in SHR than in WKY rats. Administration of U50, 448H in SHR not only caused significantly greater effects in increasing urine volume and decreasing plasma anti-diuretic hormone than in WKY rats, but also caused significant reduction in plasma angiotensin. Moreover, vasodilatory effect of U50, 488H was significantly exhibited in the renal artery segments isolated from SHR. All effects described above were abolished by nor-binaltorphimine. CONCLUSIONS: These data indicate that the depressor effect of U50, 488H in SHR is significantly stronger than that in WKY rats, and the effect is mediated or modulated by a kappa-opioid receptor sensitive mechanism. The sensitized hypotensive effect of U50, 488H in SHR may be attributed, in part, to its vasodilatory effect, enhanced beneficial effect on plasma humoral factors, and stronger diuretic effect in these hypertensive animals.


Subject(s)
3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/administration & dosage , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/pharmacology , Hypertension/drug therapy , Receptors, Opioid, kappa/agonists , Angiotensins/blood , Animals , Blood Pressure/drug effects , Diuresis/drug effects , Hypertension/etiology , Hypertension/physiopathology , In Vitro Techniques , Male , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, Opioid, kappa/physiology , Renal Artery/drug effects , Urodynamics/drug effects , Vasodilation/drug effects , Vasopressins/blood
19.
Respir Physiol Neurobiol ; 157(2-3): 187-95, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17321804

ABSTRACT

PC12 cell line has been widely used in a diverse array of neurophysiological studies including in exploration of oxygen-sensing mechanism. In present study, we first identified with immunocytochemistry and Western blot methods that interleukin-1 receptor type I was expressed in the PC12 cells. We then demonstrated with patch clamping technique that extracellular application of IL-1beta dose-dependently inhibited the outward voltage-dependent and TEA-sensitive potassium currents (I(K)) in the PC12 cells, and pre-incubation with the interleukin-1 receptor antagonist almost completely abolished this inhibitory effect. In addition, application of IL-1beta shifted steady-state inactivation of I(K) in hyperpolarizing direction, but did not alter its steady-state activation. Furthermore, IL-1beta-induced inhibition of I(K) led to a membrane depolarization and a transient increase of [Ca(2+)](i) in PC12 cells. Taking together, the present study elucidates that PC12 cells bear interleukin-1 receptor and response to IL-1beta stimulation.


Subject(s)
Gene Expression/drug effects , Interleukin-1beta/pharmacology , Receptors, Interleukin-1 Type I/metabolism , Analysis of Variance , Animals , Calcium/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Interleukin 1 Receptor Antagonist Protein/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , PC12 Cells/drug effects , PC12 Cells/metabolism , Patch-Clamp Techniques/methods , Rats , Tetraethylammonium/pharmacology
20.
Sci Rep ; 7(1): 10099, 2017 08 30.
Article in English | MEDLINE | ID: mdl-28855643

ABSTRACT

An innovative natriuretic peptide analog named CNAAC (structurally consisting of the C-terminus and ring of ANP and the N-terminus of CNP) that has been shown to exhibit potent vasodilatory, diuretic, and hypotensive effects in our previous study was evaluated for the treatment of left ventricular dysfunction following myocardial infarction. The temporal relaxation effect and metabolic status of CNAAC were determined. A myocardial ischemic model was established. Rats were randomly divided into Sham, MI, MI-ANP, MI-CNP, MI-VNP, and MI-CNAAC groups. Humoral factors were measured; echocardiography and hemodynamics methods were employed to assess the cardiac function at the fourth week after modeling. The results showed that CNAAC had a potent relaxant effect and longer duration of action than ANP, CNP, or VNP. The stability of CNAAC in blood was higher than other three NPs. Four weeks of NP administration ameliorated diastolic and systolic dysfunction, the hypertrophic index, myocardial fibrosis, and infarct size; it also restored the abnormal changes in humoral factors. These results demonstrate that CNAAC has a potent cardioprotective effect against left ventricular dysfunction after myocardial infarction. The results may lay the foundation for the clinical application of this newly designed NP chimera in the treatment and prevention of heart failure.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Cardiotonic Agents/pharmacology , Myocardial Infarction/complications , Natriuretic Peptide, C-Type/pharmacology , Recombinant Fusion Proteins/pharmacology , Ventricular Dysfunction, Left/drug therapy , Animals , Aorta, Abdominal/drug effects , Atrial Natriuretic Factor/blood , Echocardiography , Hemodynamics , Male , Natriuretic Peptide, C-Type/blood , Natriuretic Peptides , Rats, Sprague-Dawley , Recombinant Fusion Proteins/blood , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Ventricular Dysfunction, Left/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL