Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Shock ; 60(2): 255-261, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37278996

ABSTRACT

ABSTRACT: Mitochondrial damage is an important cause of heart dysfunction after severe burn injury. However, the pathophysiological process remains unclear. This study aims to examine the mitochondrial dynamics in the heart and the role of µ-calpain, a cysteine protease, in this scenario. Rats were subjected to severe burn injury treatment, and the calpain inhibitor MDL28170 was administered intravenously 1 h before or after burn injury. Rats in the burn group displayed weakened heart performance and decreased mean arterial pressure, which was accompanied by a diminishment of mitochondrial function. The animals also exhibited higher levels of calpain in mitochondria, as reflected by immunofluorescence staining and activity tests. In contrast, treatment with MDL28170 before any severe burn diminished these responses to a severe burn. Burn injury decreased the abundance of mitochondria and resulted in a lower percentage of small mitochondria and a higher percentage of large mitochondria. Furthermore, burn injury caused an increase in the fission protein DRP1 in the mitochondria and a decrease in the inner membrane fusion protein OPA1. Similarly, these alterations were also blocked by MDL28170. Of note, inhibition of calpain yielded the emergence of more elongated mitochondria along with membrane invagination in the middle of the longitude, which is an indicator of the fission process. Finally, MDL28170, administered 1 h after burn injury, preserved mitochondrial function and heart performance, and increased the survival rate. Overall, these results provided the first evidence that mitochondrial recruitment of calpain confers heart dysfunction after severe burn injury, which involves aberrant mitochondrial dynamics.


Subject(s)
Burns , Calpain , Rats , Animals , Mitochondrial Dynamics , Mitochondria/metabolism , Burns/complications , Burns/drug therapy , Burns/metabolism
2.
Cell Death Discov ; 8(1): 10, 2022 Jan 10.
Article in English | MEDLINE | ID: mdl-35013173

ABSTRACT

Stress cardiomyopathy is a major clinical complication after severe burn. Multiple upstream initiators have been identified; however, the downstream targets are not fully understood. This study assessed the role of the plasma membrane in this process and its relationship with the protease µ-calpain and tumor necrosis factor-alpha (TNF-α). Here, third-degree burn injury of approximately 40% of the total body surface area was established in rats. Plasma levels of LDH and cTnI and cardiac cell apoptosis increased at 0.5 h post burn, reached a peak at 6 h, and gradually declined at 24 h. This effect correlated well with not only the disruption of cytoskeletal proteins, including dystrophin and ankyrin-B, but also with the activation of µ-calpain, as indicated by the cleaved fragments of α-spectrin and membrane recruitment of the catalytic subunit CAPN1. More importantly, these alterations were diminished by blocking calpain activity with MDL28170. Burn injury markedly increased the cellular uptake of Evans blue, indicating membrane integrity disruption, and this effect was also reversed by MDL28170. Compared with those in the control group, cardiac cells in the burn plasma-treated group were more prone to damage, as indicated by a marked decrease in cell viability and increases in LDH release and apoptosis. Of note, these alterations were mitigated by CAPN1 siRNA. Moreover, after neutralizing TNF-α with rhTNFR:Fc, calpain activity was blocked, and heart function was improved. In conclusion, we identified µ-calpain as a trigger for severe burn-induced membrane disruption in the heart and provided evidence for the application of rhTNFR:Fc to inhibit calpain for cardioprotection.

3.
Cell Death Dis ; 11(5): 388, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32439852

ABSTRACT

Previous studies indicated that Ca2+/calmodulin-dependent kinase II (CaMKII), a kinase involved in the modulation of ryanodine receptor activity, activates Ca2+-regulated protease µ-calpain to promote myocardial ischemia/reperfusion injury. This study was performed to explore the underlying mechanisms in CaMKII-induced calpain activation to better understand heart injury. To examine the Ca2+ paradox and ischemia/reperfusion injury, isolated rat hearts were subjected to a Ca2+-free solution for 3 min, or left coronary artery occlusion for 40 min, prior to restoration of normal perfusion. Blockade of trans-sarcoplasmic reticulum Ca2+ flux using ryanodine and thapsigargin failed to prevent Ca2+ paradox-induced heart injury. In contrast, the Ca2+ paradox increased CaMKII auto-phosphorylation at Thr287, while the CaMKII inhibitor KN-62 and the Na+/Ca2+ exchanger inhibitor KB-R7943 alleviated heart injury and calpain activity. Intriguingly, the binding of µ-calpain large subunit calpain-1 (CAPN1) to phospho-CaMKII was blunted by both inhibitors. Thus, a Ca2+ leak via the ryanodine receptor is not an essential element in CaMKII-elicited calpain activation. In hearts receiving vector injection, ischemia/reperfusion caused elevated calpain activity and α-fodrin degradation, along with membrane integrity damage, similar to the effects noted in control hearts. Importantly, all these alterations were diminished with delivery of adeno-associated virus expressing mutant CaMKIIδC T287A. Ischemia/reperfusion increased CaMKII auto-phosphorylation and binding of CAPN1 to phospho-CaMKII, and facilitated the translocation of phospho-CaMKII and CAPN1 to the plasma membrane, all of which were reversed by injecting CaMKII mutant. Furthermore, the relocation capacity and the interaction of CaMKII with CAPN1 appeared to be dependent upon CaMKII autophosphorylation, as its mutant delivery increased the level of CaMKII, but did not increase membrane content of CaMKII and CAPN1, or their interactions. Together, CaMKII/calpain interaction represents a new avenue for mediating myocardial ischemia/reperfusion injury, and CaMKII likely serves as both a kinase and a carrier, thereby promoting calpain membrane translocation and activation.


Subject(s)
Calcium/metabolism , Calpain/metabolism , Ischemia/drug therapy , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Calcium-Binding Proteins/metabolism , Ischemia/metabolism , Male , Rats, Sprague-Dawley , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Thapsigargin/pharmacology
4.
Mol Med Rep ; 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31432114

ABSTRACT

Contracture or diastolic dysfunction is a primary cause of injury following ischemia/reperfusion (IR). The present study examined whether Ca2+/calmodulin­dependent kinase II (CaMKII) is involved in contracture. Isolated rat hearts were subjected to either global IR or Ca2+ paradox (CaP), which is characterized by contracture. Left ventricular end­diastolic pressure, electron microscopy and troponin I  TnI) in coronary effluent were examined to indicate the extent of contracture. Compared with control hearts, both the IR and CaP groups exhibited an increase in necrosis and apoptosis, and a marked depression in contractile function. Western blot analysis showed that IR stimulated the phosphorylation (Thr287) and oxidation (Met281/282) of CaMKII, and the phosphorylation of phospholamban (PLN), a substrate of CaMKII. By contrast, only the phosphorylation of CaMKII was increased in the CaP group. Treatment with either 3 µM KN­62, an inhibitor of CaMKII, or 5 µM KB­R7943, an inhibitor of the Na+/Ca2+ exchanger, mitigated the damage and the post­translational modification of both CaMKII and PLN. Similar to the effect of the negative inotropic agent 2,3­butanedione­monoxime, the increased cell survival after treatment with KN­62 was associated with improved diastolic function. Examination using electron microscopy and a biochemical test showed the development of contraction bands, disruption of the sarcolemmal membrane and an increase in the release of TnI in both IR and CaP hearts; these results indicated the occurrence of contracture. Furthermore, these changes were inhibited by either KN­62 or KB­R7943. Taken together, these data provided evidence that CaMKII mediates reperfusion­elicited contracture, and that the activation of CaMKII via phosphorylation is involved in this process.

5.
Burns Trauma ; 6: 28, 2018.
Article in English | MEDLINE | ID: mdl-30338266

ABSTRACT

BACKGROUND: The molecular pattern of severe burn-induced acute lung injury, characterized by cell structure damage and leukocyte infiltration, remains unknown. This study aimed to determine whether calpain, a protease involved in both processes, mediates severe burn-induced acute lung injury. METHODS: Rats received full-thickness scald burns covering 30% of the total body surface area, followed by instant fluid resuscitation. MDL28170 (Tocris Bioscience), an inhibitor of calpain, was given intravenously 1 h before or after the scald burn. The histological score, wet/dry weight ratio, and caspase-3 activity were examined to evaluate the degree of lung damage. Calpain activity and its source were detected by an assay kit and immunofluorescence staining. The proteolysis of membrane skeleton proteins α-fodrin and ankyrin-B, which are substrates of calpain, was measured by Western blot. RESULTS: Time-course studies showed that tissue damage reached a peak between 1 and 6 h post-scald burn and gradually diminished at 24 h. More importantly, calpain activity reached peak levels at 1 h and was maintained until 24 h, paralleled by lung damage to some extent. Western blot showed that the levels of the proteolyzed forms of α-fodrin and ankyrin-B correlated well with the degree of damage. MDL28170 at a dose of 3 mg/kg b. w. given 1 h before burn injury not only antagonized the increase in calpain activity but also ameliorated scald burn-induced lung injury, including the degradation of α-fodrin and ankyrin-B. Immunofluorescence images revealed calpain 1 and CD45 double-positive cells in the lung tissue of rats exposed to scald burn injury, suggesting that leukocytes were a dominant source of calpain. Furthermore, this change was blocked by MDL28170. Finally, MDL28170 given at 1 h post-scald burn injury significantly ameliorated the wet/dry weight ratio compared with burn injury alone. CONCLUSIONS: Calpain, a product of infiltrating leukocytes, is a mediator of scald burn-induced acute lung injury that involves enhancement of inflammation and proteolysis of membrane skeleton proteins. Its late effects warrant further study.

6.
Biochem Biophys Res Commun ; 491(3): 687-692, 2017 09 23.
Article in English | MEDLINE | ID: mdl-28754591

ABSTRACT

Ca2+/calmodulin-dependent protein kinase II (CaMKII) has been implicated in myocardial ischemia/reperfusion (IR) injury. The aim of this study was to determine the effect of CaMKII on the damage to membrane skeleton proteins, which is an important cause of IR injury. Isolated rat hearts were subjected to 45-min global ischemia/2-h reperfusion. Both KN-62 and KN-93 were used to inhibit CaMKII. Compared with controls, the hearts in the IR group exhibited remarkable myocardial injury area, LDH release, cell apoptosis and contractile dysfunction, along with an increase in the phosphorylation of CaMKII and its substrate phospholamban. Treatment with either KN-62 or KN-93 mitigated both the heart injury and the phosphorylation of CaMKII and phospholamban. The analysis of cell skeleton proteins revealed that IR injury resulted in an increase in the 150-kDa fragments resulting from the degradation of α-fodrin and dystrophin translocating from the sarcolemmal membrane to the cytosol and a decrease in the 220-kDa isoform of ankyrin-B. As expected, Evans blue dye staining showed an increase in membrane permeability or membrane rupture in the IR group. All of these alterations were alleviated by treatment with either KN-62 or KN-93. In addition, both KN-62 and KN-93 blocked the activity and membrane recruitment of calpain, a key protease responsible for destroying cell skeleton proteins during IR injury. In conclusion, our data provide evidence that damage to membrane skeleton proteins via calpain is a destructive downstream event of CaMKII activation in the setting of myocardial IR injury.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Cytoskeletal Proteins/metabolism , Membrane Proteins/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/administration & dosage , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Animals , Benzylamines/administration & dosage , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calpain/metabolism , Enzyme Activation , Enzyme Inhibitors/administration & dosage , Heart/drug effects , In Vitro Techniques , Male , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Rats , Rats, Sprague-Dawley , Sulfonamides/administration & dosage , Treatment Outcome
7.
Clin Exp Pharmacol Physiol ; 43(10): 951-9, 2016 10.
Article in English | MEDLINE | ID: mdl-27279457

ABSTRACT

This study determined the effects of glutamate on the Ca(2+) paradoxical heart, which is a model for Ca(2+) overload-induced injury during myocardial ischaemia and reperfusion, and evaluated its effect on a known mediator of injury, calpain. An isolated rat heart was retrogradely perfused in a Langendorff apparatus. Ca(2+) paradox was elicited via perfusion with a Ca(2+) -free Krebs-Henseleit (KH) solution for 3 minutes followed by Ca(2+) -containing normal KH solution for 30 minutes. The Ca(2+) paradoxical heart exhibited almost no viable tissue on triphenyltetrazolium chloride staining and markedly increased LDH release, caspase-3 activity, cytosolic cytochrome c content, and apoptotic index. These hearts also displayed significantly increased LVEDP and a disappearance of LVDP. Glutamate (5 and 20 mmol/L) significantly alleviated Ca(2+) paradox-induced injury. In contrast, 20 mmol/L mannitol had no effect on Ca(2+) paradox. Ca(2+) paradox significantly increased the extent of the translocation of µ-calpain to the sarcolemmal membrane and the proteolysis of α-fodrin, which suggests calpain activation. Glutamate also blocked these effects. A non-selective inhibitor of glutamate transporters, dl-TBOA (10 µmol/L), had no effect on control hearts, but it reversed glutamate-induced cardioprotection and reduction in calpain activity. Glutamate treatment significantly increased intracellular glutamate content in the Ca(2+) paradoxical heart, which was also blocked by dl-TBOA. We conclude that glutamate protects the heart against Ca(2+) overload-induced injury via glutamate transporters, and the inhibition of calpain activity is involved in this process.


Subject(s)
Calcium/toxicity , Calpain/antagonists & inhibitors , Calpain/metabolism , Cardiotonic Agents/therapeutic use , Glutamic Acid/therapeutic use , Myocardial Reperfusion Injury/metabolism , Animals , Calcium/metabolism , Cardiotonic Agents/pharmacology , Glutamic Acid/pharmacology , Heart/drug effects , Heart/physiology , Male , Myocardial Reperfusion Injury/chemically induced , Myocardial Reperfusion Injury/prevention & control , Organ Culture Techniques , Rats , Rats, Sprague-Dawley
8.
Nan Fang Yi Ke Da Xue Xue Bao ; 36(5): 633-8, 2016 May.
Article in Chinese | MEDLINE | ID: mdl-27222176

ABSTRACT

OBJECTIVE: To investigate the Effect of 2,3-butanedione monoxime (BDM) on calcium paradox-induced heart injury and its underlying mechanisms. METHODS: Thirty-two adult male SD rats were randomized into 4 groups, namely the control group, BDM treatment control group, calcium paradox group, and BDM treatment group. Isolated Sprague Dawley male rat hearts underwent Langendorff perfusion and the left ventricular pressure (LVP) and left ventricular end-diastolic pressure (LVEDP) were monitored. Left ventricular developed pressure (LVDP) was calculated to evaluate the myocardial performance. Lactate dehydrogenase (LDH) content in the coronary flow was determined. Triphenyltetrazolium chloride staining was used to measure the infarct size, and myocardial cell apoptosis was tested with TUNEL method. Western blotting was used to determine the expression of cleaved caspase-3 and cytochrome c. RESULTS: Compared with the control group, BDM at 20 mmol/L had no effect on cardiac performance, cell death, apoptotic index or the content of LDH, cleaved caspase-3 and cytochrome c at the end of perfusion under control conditions (P>0.05). Calcium paradox treatment significantly decreased the cardiac function and the level of LVDP and induced a larger infarct size (P<0.01), an increased myocardial apoptosis index (P<0.01), and up-regulated expressions of cleaved caspase-3 and cytochrome c (P<0.01). BDM (20 mmol/L) significantly attenuated these effects induced by calcium paradox, and markedly down-regulated the levels of LVEDP and LDH (P<0.01), lowered myocardial apoptosis index, decreased the content of cleaved caspase-3 and cytochrome c (P<0.01), increased LVDP, and reduced the infarct size (P<0.01). CONCLUSION: BDM suppresses cell apoptosis and contracture and improves heart function and cell survival in rat hearts exposed to calcium paradox, suggesting the value of BDM as an potential drug for myocardial ischemia reperfusion injur.


Subject(s)
Calcium/adverse effects , Diacetyl/analogs & derivatives , Myocardial Reperfusion Injury/drug therapy , Animals , Apoptosis , Caspase 3/metabolism , Cytochromes c/metabolism , Diacetyl/pharmacology , Heart/drug effects , Heart/physiopathology , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , Male , Myocardial Reperfusion Injury/chemically induced , Rats , Rats, Sprague-Dawley , Ventricular Function, Left
9.
Int J Clin Exp Pathol ; 8(9): 10239-49, 2015.
Article in English | MEDLINE | ID: mdl-26617732

ABSTRACT

BACKGROUND: Reverse-mode of the Na(+)/Ca(2+) exchanger (NCX) stimulation provides cardioprotective effects for the ischemic/reperfused heart during ischemic preconditioning (IP). This study was designed to test the hypothesis that pretreatment with an inhibitor of cardiac delayed-rectifying K(+) channel (IKr), E4031, increases reverse-mode of NCX activity, and triggers preconditioning against infarct size (IS) and arrhythmias caused by ischemia/reperfusion injury through mitoKCa channels. MATERIALS AND METHODS: In the isolated perfused rat heart, myocardial ischemia/reperfusion injury was created by occlusion of the left anterior descending coronary artery for 30 min followed by 120 min reperfusion. Two cycles of coronary occlusion for 5 min and reperfusion were performed, or pretreatment with E4031 or sevoflurane (Sevo) before the 30 min occlusion with the reversed-mode of NCX inhibitor (KB-R7943) or not. RESULTS: E4031 or Sevo preconditioning not only markedly decreased IS but also reduced arrhythmias, which was significantly blunted by KB-R7943. Furthermore, these effects of E4031 preconditioning on IS and arrhythmias were abolished by inhibition of the mitoKCa channels. Similarly, pretreatment with NS1619, an opener of the mitoKCa channels, for 10 min before occlusion reduced both the infarct size and arrhythmias caused by ischemia/reperfusion. However, these effects weren't affected by blockade of the NCX with KB-R7943. CONCLUSION: Taken together, these preliminary results conclude that pretreatment with E4031 reduces infarct size and produces anti-arrhythmic effect via stimulating the reverse-mode NCX, and that the mitoKCa channels mediate the protective effects.


Subject(s)
Ischemic Preconditioning , Mitochondria/metabolism , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Potassium Channels, Calcium-Activated/metabolism , Sodium-Calcium Exchanger/metabolism , Animals , Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/metabolism , Male , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Piperidines/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley
10.
Cell Physiol Biochem ; 35(6): 2121-34, 2015.
Article in English | MEDLINE | ID: mdl-25896056

ABSTRACT

AIMS: The aim of this study was to determine whether calpain is involved in Cl(-)-induced myocardial ischemia/reperfusion (I/R) injury. METHODS: Isolated rat hearts were subjected to either 45 min of global no-flow ischemia followed by reperfusion or successive perfusion with Ca(2+)-free KH solution for 3 min and normal KH solution for 30 min, also known as Ca(2+) paradox. RESULTS: The hearts in the I/R group exhibited increases in myocardial injury area, LDH release, caspase 3 activity and apoptotic indices and a marked decline in cardiac performance. As was the case regarding the effects of MDL 28170, an inhibitor of calpain, treatment with 5 µM NPPB, 5 µM DIDS and low Cl(-) significantly attenuated cardiac injury. Moreover, each of the treatments significantly protected against Ca(2+) overload-induced injury in the setting of Ca(2+) paradox. The Western blot and immunofluorescence data revealed that there was an increase in the percentages of calpain membrane-positive cells and the numbers of fragments resulting from the calpain-mediated proteolysis of α-fodrin in both the I/R and the Ca(2+) paradox, indicating that the activation of calpain occurred. More importantly, these effects were mitigated by the blockade of transmembrane Cl(-) flux, as was accomplished via MDL 28170. CONCLUSION: Our results provide evidence that the blockade of transmembrane Cl(-) flux mitigates I/R-induced cardiac injury via the inhibition of calpain activity. They also indicate that intracellular Ca(2+) overload regulates calpain activation in the setting of Cl(-)-induced injury.


Subject(s)
Calpain/antagonists & inhibitors , Chlorides/adverse effects , Chlorides/antagonists & inhibitors , Heart Injuries/chemically induced , Myocardial Reperfusion Injury/chemically induced , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Calcium/metabolism , Carrier Proteins/metabolism , Caspase 3/metabolism , Dipeptides/pharmacology , Heart/drug effects , Heart/physiopathology , Heart Injuries/drug therapy , Male , Microfilament Proteins/metabolism , Myocardial Reperfusion Injury/drug therapy , Proteolysis/drug effects , Rats , Rats, Sprague-Dawley
11.
PLoS One ; 8(8): e70990, 2013.
Article in English | MEDLINE | ID: mdl-23976970

ABSTRACT

BACKGROUND: Peptidylprolyl cis/trans isomerase NIMA-interacting 1 (PIN1) is involved in the process of tumorigenesis. The two single nucleotide polymorphisms (-677T>C, -842G>C) in the PIN1 promoter region have been suspected of being associated with cancer risk for years, but the conclusion is still inconclusive. METHODS: Eligible case-control studies were retrieved by searching databases and references of related reviews and studies. Genotype distribution data, adjusted odds ratios (ORs) and 95% confidence (CIs) intervals were extracted to calculate pooled ORs. RESULTS: A total of 4619 cancer cases and 4661 controls were included in this meta-analysis. Overall, the PIN1 -667T>C polymorphism was not associated with cancer risk, while the -842C allele was significantly associated with reduced cancer risk (CC+GC vs. GG, OR = 0.725, 95% CI: 0.607-0.865; P(heterogeneity) = 0.012 and GC vs. GG: OR = 0.721, 95% CI: 0.591-0.880; P(heterogeneity) = 0.003). Results from genotype distribution data were in agreement with those calculated with adjusted ORs and 95% CIs. No publication bias was detected. CONCLUSIONS: Results of this meta-analysis suggest that the PIN1 -842G>C polymorphism is associated with decreased cancer risk, but that the -667T>C polymorphism is not.


Subject(s)
Genetic Predisposition to Disease , Neoplasms/genetics , Peptidylprolyl Isomerase/genetics , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Alleles , Asian People , Case-Control Studies , Female , Genetic Association Studies , Humans , Male , NIMA-Interacting Peptidylprolyl Isomerase , Neoplasms/ethnology , Odds Ratio , Risk , White People
13.
Clin Exp Pharmacol Physiol ; 39(4): 385-92, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22356295

ABSTRACT

The calcium paradox represents an important model in which to study myocardial injuries due to intracellular Ca(2+) overload. In a previous study, calpain was transiently activated in Ca(2+) -paradoxic hearts. The aim of the present study was to determine the role of calpain in myocardial dysfunction in hearts subjected to the Ca(2+) paradox and to elucidate the underlying mechanisms. Rat hearts were isolated, Langendorff perfused and subjected to the Ca(2+) paradox, which was induced by 3 min Ca(2+) depletion followed by 30 min Ca(2+) repletion, in the presence or absence of the calpain inhibitor 10 umol/L MDL 28170. Cardiac function was evaluated. Furthermore, cell death and the degradation of troponin I (TnI) were assessed and calpain activity was determined by measurement of the α-fodrin fragment and confocal image analysis. Upon Ca(2+) repletion, the hearts immediately deteriorated, exhibiting a marked depression in cardiac function and an enlarged myocardial injury area. This was accompanied by significant increases in lactate dehydrogenase, mitochondrial release of cytochrome c, the apoptotic index and degraded TnI. These changes were significantly inhibited by MDL 28170, with the exception of TnI degradation. Compared with the control group, Ca(2+) -paradoxic hearts showed a marked increase in cleaved 150 kDa fragments resulting from specific calpain-mediated proteolysis of α-fodrin. This effect was attenuated by MDL 28170. Confocal image analysis revealed the translocation of both µ- and m-calpain to the sarcolemmal membrane in Ca(2+) -paradoxic hearts, indicating increased activity of both isoforms. The results suggest that the Ca(2+) paradox promotes calpain activity, leading to necrosis, apoptosis and myocardial dysfunction.


Subject(s)
Calcium/deficiency , Calpain/antagonists & inhibitors , Cardiotonic Agents/pharmacology , Dipeptides/pharmacology , Glycoproteins/pharmacology , Myocardium/metabolism , Animals , Calcium/pharmacology , Calpain/metabolism , Cell Death/drug effects , Cell Death/physiology , Male , Myocardium/pathology , Random Allocation , Rats , Rats, Sprague-Dawley
14.
PLoS One ; 7(12): e52270, 2012.
Article in English | MEDLINE | ID: mdl-23284963

ABSTRACT

The Ca(2+) paradox represents a good model to study Ca(2+) overload injury in ischemic heart diseases. We and others have demonstrated that contracture and calpain are involved in the Ca(2+) paradox-induced injury. This study aimed to elucidate their roles in this model. The Ca(2+) paradox was elicited by perfusing isolated rat hearts with Ca(2+)-free KH media for 3 min or 5 min followed by 30 min of Ca(2+) repletion. The LVDP was measured to reflect contractile function, and the LVEDP was measured to indicate contracture. TTC staining and the quantification of LDH release were used to define cell death. Calpain activity and troponin I release were measured after Ca(2+) repletion. Ca(2+) repletion of the once 3-min Ca(2+) depleted hearts resulted in almost no viable tissues and the disappearance of contractile function. Compared to the effects of the calpain inhibitor MDL28170, KB-R7943, an inhibitor of the Na(+)/Ca(2+) exchanger, reduced the LVEDP level to a greater extent, which was well correlated with improved contractile function recovery and tissue survival. The depletion of Ca(2+) for 5 min had the same effects on injury as the 3-min Ca(2+) depletion, except that the LVEDP in the 5-min Ca(2+) depletion group was lower than the level in the 3-min Ca(2+) depletion group. KB-R7943 failed to reduce the level of LVEDP, with no improvement in the LVDP recovery in the hearts subjected to the 5-min Ca(2+) depletion treatment; however, KB-R7943 preserved its protective effects in surviving tissue. Both KB-R7943 and MDL28170 attenuated the Ca(2+) repletion-induced increase in calpain activity in 3 min or 5 min Ca(2+) depleted hearts. However, only KB-R7943 reduced the release of troponin I from the Ca(2+) paradoxic heart. These results provide evidence suggesting that contracture is the main cause for contractile dysfunction, while activation of calpain mediates cell death in the Ca(2+) paradox.


Subject(s)
Calcium/metabolism , Calpain/metabolism , Contracture/physiopathology , Heart Injuries/physiopathology , Animals , Blotting, Western , Calpain/antagonists & inhibitors , Dipeptides/pharmacology , Heart Injuries/metabolism , Male , Myocardial Infarction , Myocardial Reperfusion Injury , Rats , Rats, Sprague-Dawley , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Calcium Exchanger/metabolism , Thiourea/analogs & derivatives , Thiourea/pharmacology
15.
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
16.
J Gen Physiol ; 135(3): 229-45, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20142516

ABSTRACT

Rapid chloride permeation through the cystic fibrosis transmembrane conductance regulator (CFTR) Cl(-) channel is dependent on the presence of fixed positive charges in the permeation pathway. Here, we use site-directed mutagenesis and patch clamp recording to show that the functional role played by one such positive charge (K95) in the inner vestibule of the pore can be "transplanted" to a residue in a different transmembrane (TM) region (S1141). Thus, the mutant channel K95S/S1141K showed Cl(-) conductance and open-channel blocker interactions similar to those of wild-type CFTR, thereby "rescuing" the effects of the charge-neutralizing K95S mutation. Furthermore, the function of K95C/S1141C, but not K95C or S1141C, was inhibited by the oxidizing agent copper(II)-o-phenanthroline, and this inhibition was reversed by the reducing agent dithiothreitol, suggesting disulfide bond formation between these two introduced cysteine side chains. These results suggest that the amino acid side chains of K95 (in TM1) and S1141 (in TM12) are functionally interchangeable and located closely together in the inner vestibule of the pore. This allowed us to investigate the functional effects of increasing the number of fixed positive charges in this vestibule from one (in wild type) to two (in the S1141K mutant). The S1141K mutant had similar Cl(-) conductance as wild type, but increased susceptibility to channel block by cytoplasmic anions including adenosine triphosphate, pyrophosphate, 5-nitro-2-(3-phenylpropylamino)benzoic acid, and Pt(NO(2))(4)(2-) in inside-out membrane patches. Furthermore, in cell-attached patch recordings, apparent voltage-dependent channel block by cytosolic anions was strengthened by the S1141K mutation. Thus, the Cl(-) channel function of CFTR is maximal with a single fixed positive charge in this part of the inner vestibule of the pore, and increasing the number of such charges to two causes a net decrease in overall Cl(-) transport through a combination of failure to increase Cl(-) conductance and increased susceptibility to channel block by cytosolic substances.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Ion Channel Gating/physiology , Membrane Potentials/physiology , Cations , Humans , Mutagenesis, Site-Directed , Patch-Clamp Techniques , Protein Conformation , Structure-Activity Relationship
17.
Int J Cardiol ; 140(2): 161-8, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-19059659

ABSTRACT

BACKGROUND: The effects of low K(+) on post-ischemic reperfused heart cells are not clearly understood. Calcium overload is one of the major causes for myocardial reperfusion injury, the present study was to investigate the role of intracellular calcium oscillations in the effects of reperfusion with low K(+) on rat myocytes. METHODS: Ischemic myocytes were reperfused with Tyrode solution containing K(+) at 5.4 (control) or 3.0 mM (low K(+)) for 10 min. the changes of intracellular calcium was recorded by spectrofluorometry. The exclusion of trypan blue by myocytes served as indices of viability. Measurements of cell length, reverse-mode Na(+)-Ca(2+) exchanger (NCX) and Na(+), K(+) ATPase activity were performed. RESULTS: Compared to control, myocytes reperfused with low K(+) had greater number of calcium oscillations and reverse-mode NCX activity, which were accompanied with decreased cell length recovery and cell viability. Reperfusion with KB-R7943, an inhibitor of reverse-mode NCX, attenuated the effects of low K(+) on all the parameters. Inhibition of Na(+), K(+) ATPase with Ouabain increased the susceptibility to calcium oscillations in myocytes reperfused with low K(+), which was accompanied with cell length shortening and decreased cell viability. Reperfusion with K(+) at 9.0 mM, which activated Na(+), K(+) ATPase, attenuated calcium oscillations, protected cell length recovery, and increased cell viability. CONCLUSIONS: These results suggest that increased calcium oscillations mediate the exacerbating reperfusion injury with low K(+) on myocytes, and inhibition of Na(+), K(+) ATPase activity and increase of reverse-mode NCX activity contribute to these effects.


Subject(s)
Calcium Signaling/physiology , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Potassium Chloride/pharmacology , Sodium-Calcium Exchanger/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Heart Ventricles/cytology , Male , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Ouabain/pharmacology , Rats , Rats, Sprague-Dawley , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors
18.
Can J Physiol Pharmacol ; 87(5): 387-95, 2009 May.
Article in English | MEDLINE | ID: mdl-19448737

ABSTRACT

Extracellular anions enter into the pore of the cystic fibrosis transmembrane conductance regulator (CFTR) Cl- channel, interacting with binding sites on the pore walls and with other anions inside the pore. There is increasing evidence that extracellular anions may also interact with sites away from the channel pore to influence channel properties. We have used site-directed mutagenesis and patch-clamp recording to identify residues that influence interactions with external anions. Anion interactions were assessed by the ability of extracellular Pt(NO2)42- ions to weaken the pore-blocking effect of intracellular Pt(NO2)42- ions, a long-range ion-ion interaction that does not appear to reflect ion interactions inside the pore. We found that mutations that remove positive charges in the 4th extracellular loop of CFTR (K892Q and R899Q) significantly alter the interaction between extracellular and intracellular Pt(NO2)42- ions. These mutations do not affect unitary Cl- conductance or block of single-channel currents by extracellular Pt(NO2)42- ions, however, suggesting that the mutated residues are not in the channel pore region. These results suggest that extracellular anions can regulate CFTR pore properties by binding to a site outside the pore region, probably by a long-range conformational change. Our findings also point to a novel function of the long 4th extracellular loop of the CFTR protein in sensing and (or) responding to anions in the extracellular solution.


Subject(s)
Anions/metabolism , Chloride Channels/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Animals , Binding Sites , Chloride Channels/physiology , Chlorides/metabolism , Cricetinae , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Mutagenesis, Site-Directed , Structure-Activity Relationship
19.
Eur J Pharmacol ; 607(1-3): 135-42, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19233160

ABSTRACT

The aim of the present study was to determine whether U50,488H (a selective kappa-opioid receptor agonist) inhibits cardiac hypertrophy and fibrosis induced by beta-adrenoceptor stimulation in a rat model. Cardiac hypertrophy and fibrosis were developed by intraperitoneal administration of isoprenaline (ip. 3.0 mg/kg/day,14 days). In the isoprenaline-treated group, heart weight and heart-to-body ratio increased significantly. Hypertrophic alterations were observed in light micrographs of tissue and transmission electron micrographs of myocardial ultra structures. Increases in heart weight, heart-to-body ratio, diameter of cardiomyocytes, and morphological hypertrophic alterations induced by isoprenaline were significantly attenuated by U50,488H(i.p. 1.25 mg/kg/day). Growth of cardiomyocytes was induced by incubating with isoprenaline (10(-6) mol/l), which resulted in an increase in optical density (OD) values. The increased OD value was attenuated by U50,488H(10(-7) mol/l-10(-5) mol/l) in a dose dependent manner. Animals receiving administration of isoprenaline displayed significant fibrosis. The extent of isoprenaline induced left ventricular fibrosis was dramatically reduced in U50,488H treated animals. Increased cardiac fibroblast proliferation and collagen synthesis induced by isoprenaline, as evidenced by increased OD value, (3)H-thymidine, and (3)H-proline incorporation, were significantly reduced in the U50,488H treated group. The specific extracellular matrix proteins, including type I, type III collagen and fibronectin, which increased after administration of isoproterenol, were also attenuated by U50,488H. The abovementioned effects of U50,488H were completely abolished by nor-BNI (nor-binaltorphimine), a selective kappa-opioid receptor antagonist. The enhanced intracellular Ca(2+) transient and L-type Ca(2+) current elicited by isoprenaline in cardiomyocytes were significantly inhibited by U50,488H. This study provides the first morphological evidence of the inhibitory effect of U50,488H on isoprenaline-induced cardiac hypertrophy and fibrosis via kappa-opioid receptor stimulation.


Subject(s)
3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Cardiomegaly/prevention & control , Fibrosis/prevention & control , Receptors, Opioid, kappa/agonists , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/administration & dosage , Animals , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/pharmacology , Calcium/metabolism , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/metabolism , Cardiomegaly/physiopathology , Collagen Type I/drug effects , Collagen Type I/metabolism , Collagen Type III/drug effects , Collagen Type III/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Fibronectins/drug effects , Fibronectins/metabolism , Fibrosis/physiopathology , Heart Ventricles/drug effects , Heart Ventricles/pathology , Isoproterenol , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley
20.
J Thorac Cardiovasc Surg ; 136(6): 1450-5, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19114188

ABSTRACT

OBJECTIVES: We sought to evaluate a moderate-potassium cardioplegic solution using adenosine and lidocaine as the arresting and protecting cardioprotective combination in pediatric cardiac surgery. METHODS: One hundred thirty-four patients with congenital heart disease were randomly allocated to one of 3 groups according to the cardioplegia formula used: the high-potassium (HP) group (K(+), 20 mmol/L), 46 patients; the high-potassium adenosine-lidocaine (HPAL) group (K(+), 20 mmol/L; adenosine, 0.7 mmol/L; and lidocaine, 0.7 mmol/L), 44 patients; and the moderate-potassium adenosine-lidocaine (MPAL) group (K(+), 10 mmol/L; adenosine, 0.7 mmol/L; and lidocaine, 0.7 mmol/L), 44 patients. Hemodynamic data during the operation and postoperative data were recorded. Serum cardiac troponin I concentrations were examined at the time points of before cardiopulmonary bypass and 1, 3, 6, 12, and 24 hours after aortic crossclamp removal. RESULTS: At the end of cardiopulmonary bypass and modified ultrafiltration, the systolic and pulse pressures of the MPAL group were significantly increased compared with the respective values of the HP group. At the time points of 1 to 12 hours after reperfusion, the levels of serum cardiac troponin I were significantly decreased in the MPAL group compared with those in the HP and HPAL groups. CONCLUSIONS: The MPAL cardioplegia formula was associated with better myocardial protective effects.


Subject(s)
Cardioplegic Solutions/pharmacology , Cardiotonic Agents/pharmacology , Heart Defects, Congenital/surgery , Heart/drug effects , Adenosine/administration & dosage , Adenosine/pharmacology , Cardiac Surgical Procedures , Cardioplegic Solutions/administration & dosage , Cardiopulmonary Bypass , Cardiotonic Agents/administration & dosage , Child, Preschool , Female , Humans , Infant , Lidocaine/administration & dosage , Lidocaine/pharmacology , Male , Myocardium , Potassium/administration & dosage , Potassium/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...