Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Am J Physiol Heart Circ Physiol ; 296(5): H1633-42, 2009 May.
Article in English | MEDLINE | ID: mdl-19252088

ABSTRACT

alphaB-crystallin (alphaBC) is a small heat shock protein expressed at high levels in the myocardium where it protects from ischemia-reperfusion damage. Ischemia-reperfusion activates p38 MAP kinase, leading to the phosphorylation of alphaBC on serine 59 (P-alphaBC-S59), enhancing its ability to protect myocardial cells from damage. In the heart, ischemia-reperfusion also causes the translocation of alphaBC from the cytosol to other cellular locations, one of which was recently shown to be mitochondria. However, it is not known whether alphaBC translocates to mitochondria during ischemia-reperfusion, nor is it known whether alphaBC phosphorylation takes place before or after translocation. In the present study, analyses of mitochondrial fractions isolated from mouse hearts subjected to various times of ex vivo ischemia-reperfusion showed that alphaBC translocation to mitochondria was maximal after 20 min of ischemia and then declined steadily during reperfusion. Phosphorylation of mitochondrial alphaBC was maximal after 30 min of ischemia, suggesting that at least in part it occurred after alphaBC association with mitochondria. Consistent with this was the finding that translocation of activated p38 to mitochondria was maximal after only 10 min of ischemia. The overexpression of alphaBC-AAE, which mimics alphaBC phosphorylated on serine 59, has been shown to stabilize mitochondrial membrane potential and to inhibit apoptosis. In the present study, infection of neonatal rat cardiac myocytes with adenovirus-encoded alphaBC-AAE decreased peroxide-induced mitochondrial cytochrome c release. These results suggest that during ischemia alphaBC translocates to mitochondria, where it is phosphorylated and contributes to modulating mitochondrial damage upon reperfusion.


Subject(s)
Mitochondria, Heart/metabolism , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion , alpha-Crystallin B Chain/metabolism , Animals , Animals, Newborn , Cells, Cultured , Cytochromes c/metabolism , Hydrogen Peroxide/toxicity , Kinetics , Mice , Mice, Inbred C57BL , Mitochondria, Heart/drug effects , Mitochondria, Heart/enzymology , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Phosphorylation , Protein Transport , Rats , Transduction, Genetic , alpha-Crystallin B Chain/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Am J Physiol Heart Circ Physiol ; 294(1): H337-44, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17993600

ABSTRACT

The cytosolic small heat shock protein alphaB-crystallin (alphaBC) is a molecular chaperone expressed in large quantities in the heart, where it protects from stresses such as ischemia-reperfusion (I/R). Upon I/R, p38 MAP kinase activation leads to phosphorylation of alphaBC on Ser(59) (P-alphaBC-S59), which increases its protective ability. alphaBC confers protection, in part, by interacting with and affecting the functions of key components in stressed cells. We investigated the hypothesis that protection from I/R damage in the heart by P-alphaBC-S59 can be mediated by localization to mitochondria. We found that P-alphaBC-S59 localized to mitochondria isolated from untreated mouse hearts and that this localization increased more than threefold when the hearts were subjected to ex vivo I/R. Mitochondrial P-alphaBC-S59 decreased when hearts were treated with the p38 inhibitor SB-202190. Moreover, SB-202190-treated hearts exhibited more tissue damage and less functional recovery upon reperfusion than controls. I/R activates mitochondrial permeability transition (MPT) pore opening, which increases cell damage. We found that mitochondria incubated with a recombinant mutant form of alphaBC that mimics P-alphaBC-S59 exhibited decreased calcium-induced MPT pore opening. These results indicate that mitochondria may be among the key components in stressed cells with which P-alphaBC-S59 interacts and that this localization may protect the myocardium, in part, by modulating MPT pore opening and, thus, reducing I/R injury.


Subject(s)
Mitochondria, Heart/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , alpha-Crystallin B Chain/metabolism , Animals , Calcium/metabolism , Cytosol/metabolism , Female , Imidazoles/pharmacology , In Vitro Techniques , Mice , Mice, Inbred C3H , Mitochondria, Heart/drug effects , Mitochondria, Heart/enzymology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Mutation , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/metabolism , Myocardium/enzymology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Recombinant Proteins/metabolism , Subcellular Fractions/metabolism , Time Factors , alpha-Crystallin B Chain/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Cell Death Differ ; 14(1): 146-57, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16645637

ABSTRACT

Ischemia and reperfusion (I/R) injury is associated with extensive loss of cardiac myocytes. Bnip3 is a mitochondrial pro-apoptotic Bcl-2 protein which is expressed in the adult myocardium. To investigate if Bnip3 plays a role in I/R injury, we generated a TAT-fusion protein encoding the carboxyl terminal transmembrane deletion mutant of Bnip3 (TAT-Bnip3DeltaTM) which has been shown to act as a dominant negative to block Bnip3-induced cell death. Perfusion with TAT-Bnip3DeltaTM conferred protection against I/R injury, improved cardiac function, and protected mitochondrial integrity. Moreover, Bnip3 induced extensive fragmentation of the mitochondrial network and increased autophagy in HL-1 myocytes. 3D rendering of confocal images revealed fragmented mitochondria inside autophagosomes. Enhancement of autophagy by ATG5 protected against Bnip3-mediated cell death, whereas inhibition of autophagy by ATG5K130R enhanced cell death. These results suggest that Bnip3 contributes to I/R injury which triggers a protective stress response with upregulation of autophagy and removal of damaged mitochondria.


Subject(s)
Autophagy , Membrane Proteins/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/cytology , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis , Gene Deletion , Male , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Mitochondria, Heart/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/isolation & purification , Mitochondrial Proteins/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/isolation & purification , Rats , Rats, Sprague-Dawley
4.
Cell Death Differ ; 11 Suppl 2: S144-52, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15608693

ABSTRACT

Ten years ago, the first finding of apoptotic cell death on the 'crime scene' of cardiac ischemia/reperfusion injury profoundly dismayed the scientific community. This observation jarred with the deeply rooted conviction that cardiac myocytes stoically 'break, but do not bend' in the fight against ischemia, instead of spontaneously accepting a peaceful demise for the greater good. Ten years later, a number of studies not only proved right the coexistence of necrosis and apoptosis on the ischemic battle field, but also implicated myocyte apoptosis in the pathogenesis of all the shapes and shades that cardiac ischemic injury can take on.


Subject(s)
Apoptosis/physiology , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , Animals , Cell Death/physiology , Humans , Mitochondria, Heart/physiology , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/physiopathology
5.
Am J Physiol Heart Circ Physiol ; 285(5): H2218-24, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12805018

ABSTRACT

Type II secretory phospholipase A2 (sPLA2) is a cardiovascular risk factor. We recently found depositions of sPLA2 in the necrotic center of infarcted human myocardium and normally appearing cardiomyocytes adjacent to the border zone. The consequences of binding of sPLA2 to ischemic cardiomyocytes are not known. To explore a potential effect of sPLA2 on ischemic cardiomyocytes at a cellular level we used an in vitro model. The cardiomyocyte cell line H9c2 or adult cardiomyocytes were isolated from rabbits that were incubated with sPLA2 in the presence of metabolic inhibitors to mimic ischemia-reperfusion conditions. Cell viability was established with the use of annexin V and propidium iodide or 7-aminoactinomycin D. Metabolic inhibition induced an increase of the number of flip-flopped cells, including a population that did not stain with propidium iodide and that was caspase-3 negative. sPLA2 bound to the flip-flopped cells, including those negative for caspase-3. sPLA2 binding induced cell death in these latter cells. In addition, sPLA2 potentiated the binding of C-reactive protein (CRP) to these cells. We conclude that by binding to flip-flopped cardiomyocytes, including those that are caspase-3 negative and presumably reversibly injured, sPLA2 may induce cell death and tag these cells with CRP.


Subject(s)
Cell Death/physiology , Myocardial Ischemia/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/enzymology , Phospholipases A/metabolism , Animals , C-Reactive Protein/metabolism , C-Reactive Protein/pharmacology , Calcium/metabolism , Cell Death/drug effects , Cell Line , Chelating Agents/pharmacology , Egtazic Acid/pharmacology , Energy Metabolism/drug effects , Group II Phospholipases A2 , Male , Phospholipases A/pharmacology , Phospholipases A2 , Rabbits , Rats
7.
Circ Res ; 89(5): 461-7, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11532908

ABSTRACT

The objective of this study was to identify the mitochondrial proteins that undergo changes in phosphorylation during global ischemia and reperfusion in the isolated rabbit heart. We also assessed whether the cardioprotective intervention of ischemic preconditioning affected mitochondrial protein phosphorylation. We established a reconstituted system using isolated mitochondria and cytosol from control or ischemic hearts. We found that phosphorylation of a 46-kDa protein on a serine residue was increased in ischemia and that phosphorylation was reduced in control or preconditioned hearts. Using 2D gel electrophoresis and mass spectrometry, we have identified the 46-kDa protein as mitochondrial translational elongation factor Tu (EF-Tu(mt)). These data reveal that ischemia and preconditioning modulate the phosphorylation of EF-Tu(mt) and suggest that the mitochondrial protein synthesis machinery may be regulated by phosphorylation. Phosphorylation of mitochondrial EF-Tu has not been previously described; however, in prokaryotes, EF-Tu phosphorylation inhibits protein translation. We hypothesized that phosphorylation of mitochondrial EF-Tu would inhibit mitochondrial protein translation and attempted to reproduce the effect with inhibition of mitochondrial protein synthesis by chloramphenicol. We found that chloramphenicol pretreatment significantly reduced infarct size, suggesting that mitochondrial protein synthesis is one determinant of myocardial injury during ischemia and reperfusion.


Subject(s)
Mitochondria, Heart/metabolism , Myocardial Ischemia/metabolism , Peptide Elongation Factor Tu/metabolism , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Chloramphenicol/pharmacology , Enzyme Inhibitors/pharmacology , Genistein/pharmacology , Ischemic Preconditioning, Myocardial , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Phosphorylation/drug effects , Protein Subunits , Protein Synthesis Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Rabbits
8.
Am J Physiol Gastrointest Liver Physiol ; 281(3): G681-7, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11518680

ABSTRACT

Expression of a mutated cystic fibrosis transmembrane conductance regulator (CFTR) has been shown to enhance proliferation within CF airways, and cells expressing a mutated CFTR have been shown to be less susceptible to apoptosis. Because the CFTR is expressed in the epithelial cells lining the gastrointestinal tract and all CF mouse models are characterized by gastrointestinal obstruction, we hypothesized that CFTR null mice would have increased epithelial cell proliferation and reduced apoptosis within the small intestine. The rate of intestinal epithelial cell migration from crypt to villus was increased in CFTR null mice relative to mice expressing the wild-type CFTR. This difference in migration could be explained by an increase in epithelial cell proliferation but not by a difference in apoptosis within the crypts of Lieberkühn. In addition, using two independent sets of CF cell lines, we found that epithelial cell susceptibility to apoptosis was unrelated to the presence of a functional CFTR. Thus increased proliferation but not alterations in apoptosis within epithelial cells might contribute to the pathophysiology of CF.


Subject(s)
Apoptosis , Cell Movement , Cystic Fibrosis/physiopathology , Epithelial Cells/pathology , Intestine, Small/pathology , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Bromodeoxyuridine , Cell Division/genetics , Cell Movement/genetics , Cells, Cultured , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/biosynthesis , Cystic Fibrosis Transmembrane Conductance Regulator/deficiency , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Disease Models, Animal , Dose-Response Relationship, Radiation , Epithelial Cells/metabolism , Epithelial Cells/radiation effects , Gamma Rays , Intestine, Small/radiation effects , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/radiation effects , Mice , Mice, Inbred CFTR , Ultraviolet Rays
9.
J Biol Chem ; 276(33): 30724-8, 2001 Aug 17.
Article in English | MEDLINE | ID: mdl-11404357

ABSTRACT

Reperfusion after myocardial ischemia is associated with a rapid influx of calcium, leading to activation of various enzymes including calpain. Isolated perfused adult rabbit hearts subjected to global ischemia and reperfusion were studied. Calpain or a calpain-like activity was activated within 15 min after reperfusion, and preconditioning suppressed calpain activation. In contrast, caspase activation was not detected although cytochrome c was released after ischemia and reperfusion. The pro-apoptotic BH3-only Bcl-2 family member, Bid, was cleaved during ischemia/reperfusion in the adult rabbit heart. Recombinant Bid was cleaved by calpain to a fragment that was able to mediate cytochrome c release. The calpain cleavage site was mapped to a region within Bid that is extremely susceptible to proteolysis. These findings suggest that there is cross-talk between apoptotic and necrotic pathways in myocardial ischemia/reperfusion injury.


Subject(s)
Calpain/metabolism , Carrier Proteins/metabolism , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Amino Acid Sequence , Animals , BH3 Interacting Domain Death Agonist Protein , Male , Molecular Sequence Data , Rabbits , Recombinant Proteins/metabolism
10.
Biol Signals Recept ; 10(3-4): 147-61, 2001.
Article in English | MEDLINE | ID: mdl-11351125

ABSTRACT

Apoptosis is a coordinated sequence of events culminating in the death of the cell. Many of these biochemical processes are regulated by the mitochondria, including the release of proapoptotic molecules in addition to the caspase-activating cofactor, cytochrome c. Pro- and antiapoptotic members of the Bcl-2 family regulate mitochondrial participation in cell death. Current models explaining cytochrome c release are discussed in light of mitochondrial structure and physiology.


Subject(s)
Apoptosis , Mitochondria/metabolism , Caspases/metabolism , Cytochrome c Group/metabolism , Models, Biological , Organelles/metabolism , Porins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Voltage-Dependent Anion Channels , bcl-2-Associated X Protein
11.
Antioxid Redox Signal ; 3(1): 113-23, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11294190

ABSTRACT

Protection of ischemic myocardium is an important unmet need in reperfusion therapy of acute myocardial infarction. Myocardial ischemia and reperfusion induce necrosis and apoptosis in cardiomyocytes. Caspase processing and activation are critical steps in most receptor and nonreceptor pathways of apoptosis. Caspase inhibitors have been shown to reduce ischemia reperfusion injury in cardiac muscle. Information about dose response and time of administration are needed to optimize the design of preclinical studies. We used isolated adult rabbit cardiomyocytes subjected to metabolic inhibition (MI) and recovery to examine the role of caspases and caspase inhibitors, the dose response, and the timing of administration. In vitro inhibitory concentrations (Ki) were determined for purified caspases. Cardiomyocytes subjected to MI were treated with peptidomimetic fluoromethyl ketone inhibitors of caspases before or during MI, or at recovery. Caspase inhibitors were most effective when added before MI and included throughout recovery, but were partially protective if added after MI. The optimal concentration of the inhibitors tested was approximately 10 microM. Protection was sustained when cells were allowed to recover for 4 or 24 h. These results suggest that caspase activation is an important component of myocyte injury mediated by MI and recovery. Low doses of caspase inhibitors were identified that reduce injury in this model system, and further investigations using in vivo models are warranted.


Subject(s)
Caspase Inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Heart/drug effects , Myocardium/enzymology , Animals , Apoptosis/drug effects , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , Myocardium/cytology , Rabbits , Signal Transduction
12.
Mitochondrion ; 1(1): 61-9, 2001 Jun.
Article in English | MEDLINE | ID: mdl-16120269

ABSTRACT

The mitochondria have been shown to play a key role in the initiation of caspase activation during apoptosis. Recently, some caspases have been shown to be associated with mitochondria. In this study, we used Jurkat T-lymphoblasts to show that caspases -2 and -3 are located in the mitochondrial intermembrane space, associated with the inner membrane. Caspase-9 is associated with the outer membrane and is exposed to the cytosolic compartment. Caspase activation took place predominantly in the cytosol in response to Fas ligation, but staurosporine treatment led to caspase activation in both cytosol and mitochondria. In response to both Fas and staurosporine treatment, caspase processing could be detected earlier in cytosol than in mitochondria, but this could reflect the limits of sensitive detection by immunoblotting. Only trace amounts of Apaf-1 were found in association with the mitochondria. However, staurosporine treatment led to preferential auto-processing of caspase-9 associated with mitochondria. These findings suggest that mitochondrial caspases are regulated independently of the cytosolic pool of caspases. The data are also consistent with the notion of a caspase nucleation site associated with mitochondria. Using a stable transfected CEM cell line, we show that Bcl-2 suppressed caspase processing in both cytosolic and mitochondrial compartments in response to both staurosporine and Fas ligation.

13.
FEBS Lett ; 482(1-2): 6-12, 2000 Sep 29.
Article in English | MEDLINE | ID: mdl-11018514

ABSTRACT

Mitochondria play an essential function in eukaryotic life and death. They also play a central role in apoptosis regulation, reflected by the convergence of Bcl-2 family members on the mitochondrial outer membrane, and the presence of 'death factors' in the intermembrane space. Mitochondrial structure and function must be taken into consideration when evaluating mechanisms for cytochrome c release. The core machinery for caspase activation is conserved from Caenorhabditis elegans to man, and we consider parallels in the role of mitochondria in this process.


Subject(s)
Apoptosis/physiology , Mitochondria/physiology , Animals , Caenorhabditis elegans , Caspases/metabolism , Cytochrome c Group/metabolism , Humans , Mitochondria/ultrastructure , Models, Biological , Proto-Oncogene Proteins c-bcl-2/metabolism
14.
FEBS Lett ; 478(1-2): 19-25, 2000 Jul 28.
Article in English | MEDLINE | ID: mdl-10922462

ABSTRACT

L-Carnitine facilitates the transport of fatty acids into the mitochondrial matrix where they are used for energy production. Recent studies have shown that L-carnitine is capable of protecting the heart against ischemia/reperfusion injury and has beneficial effects against Alzheimer's disease and AIDS. The mechanism of action, however, is not yet understood. In the present study, we found that in Jurkat cells, L-carnitine inhibited apoptosis induced by Fas ligation. In addition, 5 mM carnitine potently inhibited the activity of recombinant caspases 3, 7 and 8, whereas its long-chain fatty acid derivative palmitoylcarnitine stimulated the activity of all the caspases. Palmitoylcarnitine reversed the inhibition mediated by carnitine. Levels of carnitine and palmitoyl-CoA decreased significantly during Fas-mediated apoptosis, while palmitoylcarnitine formation increased. These alterations may be due to inactivation of beta-oxidation or to an increase in the activity of the enzyme that converts carnitine to palmitoylcarnitine, carnitine palmitoyltransferase I (CPT I). In support of the latter possibility, fibroblasts deficient in CPT I activity were relatively resistant to staurosporine-induced apoptosis. These observations suggest that caspase activity may be regulated in part by the balance of carnitine and palmitoylcarnitine.


Subject(s)
Apoptosis/drug effects , Carnitine/pharmacology , Caspases/metabolism , Palmitoylcarnitine/pharmacology , fas Receptor/physiology , Acylation , Carnitine/analogs & derivatives , Carnitine/antagonists & inhibitors , Carnitine/metabolism , Carnitine O-Palmitoyltransferase/deficiency , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Caspase 3 , Caspase 7 , Caspase 8 , Caspase 9 , Caspase Inhibitors , Cell Line , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Fibroblasts , Humans , Jurkat Cells , Palmitoyl Coenzyme A/metabolism , Palmitoylcarnitine/antagonists & inhibitors , Palmitoylcarnitine/metabolism , Staurosporine/pharmacology
16.
Crit Rev Eukaryot Gene Expr ; 10(3-4): 231-9, 2000.
Article in English | MEDLINE | ID: mdl-11272466

ABSTRACT

Apoptosis is characterized by biochemical processes that are largely conserved throughout evolution. The basic elements of the system comprise caspases, their activators and inhibitors, and regulators of mitochondrial integrity. New evidence reveals the role of mitochondria as the central coordinators of apoptosis. Accordingly, some caspases are sequestered within the mitochondria, and mitochondria contain additional proapoptotic factors. Bcl-2 and Bax homologs regulate the integrity of the mitochondrial outer membrane, which may also serve as a scaffold for the apoptotic machinery.


Subject(s)
Apoptosis/physiology , Mitochondria/physiology , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/enzymology , Caspases/metabolism , Cytochrome c Group/metabolism , Enzyme Activation , Mitochondria/enzymology , Mitochondria/ultrastructure , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptor Cross-Talk
17.
Drug News Perspect ; 13(8): 471-6, 2000 Oct.
Article in English | MEDLINE | ID: mdl-12937619

ABSTRACT

Developmentally programmed cell death in animals is accomplished by the activation of a protease of the caspase family. Caspase activation is an essential feature of apoptosis. In Caenorhabditis elegans, this protease is CED-3, which corresponds to mammalian caspase-3. Caspases comprise a distinct family of cysteine aspartases that are activated by interaction with a co-factor and/or proteolytic processing. Once activated, they cleave targets containing the exposed consensus sequences, including other caspases, protein kinases and structural elements, to achieve the death of the cell. Apoptotic cells undergo a dramatic volume loss accompanied by ionic shifts and cytoplasmic acidification. The cytoskeleton rearranges and the cell membrane undergoes blebbing and phosphatidylserine externalization, thus marking the dying cell for ingestion by phagocytes. In addition to structural changes, mitochondria cease to synthesize ATP, release cytochrome c and other constituents, and lose membrane potential. DNA undergoes endonucleolytic cleavage first into 50-kb fragments, followed by cleavage to oligonucleosomes. Together these biochemical processes achieve the noninflammatory destruction of the cell.

18.
Cell Death Differ ; 6(10): 987-91, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10556976

ABSTRACT

Multiple signaling pathways, including the c-Jun N-terminal kinase (JNK) pathway, are activated in myocardial ischemia and reperfusion (MI/R) and correlate with cell death. However, the role of the JNK pathway in MI/R-induced cell death is poorly understood. In a rabbit model, we found that ischemia followed by reperfusion resulted in JNK activation which could be detected in cytosol as well as in mitochondria. To address the functional role of the JNK activation, we examined the consequences of blockade of JNK activation in isolated cardiomyocytes under conditions of simulated ischemia. The JNK activity was stimulated approximately sixfold by simulated ischemia and reperfusion (simulated MI). When a dominant negative mutant of JNK kinase-2 (dnJNKK2), an upstream regulator of JNK, and JNK-interacting protein-1 (JIP-1) were expressed in myocytes by recombinant adenovirus, the activation of JNK by simulated MI was reduced 53%. Furthermore, the TNFalpha-activated JNK activity in H9c2 cells was completely abolished by dnJNKK2 and JIP-1. In correlation, when dnJNKK2 and JIP-1 were expressed in cardiomyocytes, both constructs significantly reduced cell death after simulated MI compared to vector controls. We conclude that activation of the JNK cascade is important for cardiomyocyte death in response to simulated ischemia.


Subject(s)
Carrier Proteins/metabolism , Cell Death/physiology , Heart/physiology , Mitogen-Activated Protein Kinase Kinases , Myocardial Ischemia/physiopathology , Myocardium/cytology , Protein Kinases/metabolism , Reperfusion Injury/physiopathology , Tumor Necrosis Factor-alpha/physiology , Adenoviridae , Adenoviridae Infections , Animals , Disease Models, Animal , Gene Expression Regulation, Viral , MAP Kinase Kinase 7 , Muscle Fibers, Skeletal/virology , Rabbits , Viral Proteins/physiology
20.
Ann N Y Acad Sci ; 874: 412-26, 1999 Jun 30.
Article in English | MEDLINE | ID: mdl-10415551

ABSTRACT

The signal transduction pathways by which ischemia-reperfusion leads to apoptosis may involve the JNK pathway, ceramide generation, and inhibition of protective PKC pathways. The biochemical events associated with apoptosis include mitochondrial inactivation, cytochrome c dislocation, caspase activation, and cytoplasmic acidification. Through the concerted efforts of multiple classes of enzymes, apoptosis is accomplished, resulting in the death of a cell in which potentially transforming oncogenes have been degraded and inflammatory contents are contained within the plasma membrane until the fragments can be ingested by phagocytes. This non-inflammatory mode of cell death permits tissue remodeling with minimal scar formation, and so is preferable to necrotic cell death. The distinction between apoptosis and necrosis, which implies different mechanisms of cell death, is blurred in the case of a pathologic insult such as ischemia-reperfusion. It is suggested that it is more useful to view cell death in the context of whether or not it can be prevented.


Subject(s)
Apoptosis/physiology , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/physiopathology , Animals , Heart/physiopathology , Myocardium/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...