Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Mol Cell Cardiol ; 33(9): 1713-7, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11549349

ABSTRACT

The lysophospholipids sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA) stimulate cellular proliferation and affect numerous cellular functions by signaling through G protein-coupled endothelial differentiation gene-encoded (Edg) receptors. S1P and LPA also act as survival factors in many cell types, but have not previously been studied in cardiac myocytes. We incubated neonatal rat cardiac myocytes either in room air/1% CO2 (normoxia) or in an atmosphere of 99% N2/1%CO2 (hypoxia) at 37 degrees C for 18-20 h in the absence of glucose. Cell viability was measured using a calcein ester green fluorescence assay. Under normoxic conditions 88.7+/-1.0% of the cells were viable after 18-20 h. Severe hypoxia reduced viability to 61.3+/-4.3% (n=6, P<0.05). In myocytes preincubated with either 10 microM S1P or 1 microM LPA for 2 h, the effects of severe hypoxia on cell viability were prevented resulting in survival equivalent to normoxia. Neither the protein kinase C inhibitor chelethyrine (1 microM) nor the mitochondrial K(ATP) channel antagonist 5-hydroxydecanoic acid, (5-HD, 100 microM) had any effect on myocyte survival during severe hypoxia, but both agents completely abolished the ability of S1P to rescue cardiac myocytes from hypoxic cell death. We also tested the effects of dimethylsphingosine (DMS), which inhibits sphingosine kinase synthesis of S1P. Incubation of neonatal rat cardiac myocytes with 10 microM DMS for 2 h in the presence of serum resulted in 25-30% cell death during 18-20 h of normoxia. DMS-induced cell death was prevented by concurrent preincubation with either S1P or GM-1, a ganglioside that activates sphingosine kinase to increase intracellular levels of S1P. We conclude that both S1P and LPA are cardioprotective for hypoxic neonatal rat ventricular myocytes. S1P acts through cellular membrane receptors by signaling mechanisms involving protein kinase C and mitochondrial K(ATP) channels. Both endogenous and exogenously applied S1P are effective in preventing cell death induced by inhibition of sphingosine kinase.


Subject(s)
Cardiotonic Agents/pharmacology , Cell Hypoxia/physiology , Cell Survival/drug effects , Lysophospholipids/pharmacology , Myocardium/cytology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Alkaloids , Animals , Animals, Newborn , Anti-Arrhythmia Agents/pharmacology , Benzophenanthridines , Cardiotonic Agents/metabolism , Cell Survival/physiology , Cells, Cultured , Culture Media, Serum-Free , Decanoic Acids/pharmacology , Enzyme Inhibitors/pharmacology , Gelsolin/pharmacology , Heart/drug effects , Hydroxy Acids/pharmacology , Phenanthridines/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Rats , Rats, Sprague-Dawley , Sphingosine/metabolism
2.
J Mol Cell Cardiol ; 32(10): 1779-86, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11013122

ABSTRACT

Although mouse models have been increasingly used for studies of cardiac pathophysiology, there is little information regarding cultured murine cardiac myocytes. Accordingly, we have developed a cell culture model of neonatal mouse cardiac myocytes by modifying a protocol used to prepare neonatal rat myocytes. The principal change is the substitution of cytosine arabinoside for bromodeoxyuridine to prevent fibroblast proliferation. Neonatal murine myocytes exhibited persistent spontaneous contraction and were viable for up to 14 days in culture. By flow cytometry 85% of the cells were cardiac myocytes. In sparse cultures (average cell density 259 cells/mm(2)), both hypoxic preconditioning (n=5) and phenylephrine pretreatment (n=8) produced significant protection of cardiac myocytes from cell death during a prolonged period of severe hypoxia (<0.5% O(2)for 18-20 h, both P<0.05). The phenylephrine effect was inhibited by the alpha(1)-adrenoceptor antagonist prazosin (n=4, P<0.05) and by an xi PKC peptide antagonist (xi V1-2) coupled to a TAT peptide (n=5, P<0. 05). Interestingly, the mixed alpha(1)- and beta -adrenoceptor agonist norepinephrine, which stimulates hypertrophy as measured by(14)[C]phenylalanine incorporation in neonatal rat cardiac myocytes, did not cause hypertrophy in mouse myocytes, suggesting that the signaling pathways for myocardial protection and hypertrophy are likely to be both divergent and species specific. In cardiac myocytes prepared from transgenic mice either homozygous or heterozygous for human Cu/Zn superoxide dismutase, there was protection from cell death (n=3) and restoration of(14)[C]phenyl- alanine uptake (n=4) during prolonged hypoxia (1% O(2)for 3 days, both P<0.05). We conclude that this cellular model, which is relatively simple to prepare, can be used for in-vitro examination of cardiac protection induced by preconditioning agents, various transgenes, and potentially by targeted gene deletions.


Subject(s)
Myocardium/cytology , Adrenergic alpha-Antagonists/pharmacology , Alanine/metabolism , Animals , Animals, Newborn , Bromodeoxyuridine/pharmacology , Cardiotonic Agents/pharmacology , Cell Survival/drug effects , Cells, Cultured , Cytarabine/pharmacology , Female , Fibroblasts/metabolism , Flow Cytometry , Gene Deletion , Humans , Hypoxia , Ischemic Preconditioning, Myocardial , Male , Mice , Mice, Transgenic , Myocardium/metabolism , Norepinephrine/pharmacology , Oxidative Stress , Phenylephrine/pharmacology , Prazosin/pharmacology , Recombinant Fusion Proteins/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Time Factors , Transgenes , Vasoconstrictor Agents/pharmacology
3.
J Mol Cell Cardiol ; 32(11): 2091-5, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11040112

ABSTRACT

Moderate alcohol consumption protects against coronary heart disease by unclear mechanisms. We tested whether chronic ethanol preconditioning requires activation of mitochondrial K(ATP)channels. Rats were fed 18% (v/v) ethanol in drinking water for 10 months. Blood alcohol levels at sacrifice were 3 mmol/l (0.015 gram percent). Isolated crystalloid-perfused hearts were subjected to global ischemia and reperfusion on a modified Langendorff apparatus. Prior alcohol exposure doubled the recovery of LVDP during reperfusion (45+/-5%v 20+/-3% of baseline for controls, n=6, P<0.01) and blunted the rise in LVEDP (3.5+/-0.5 v 5.5+/-0.4 times baseline for controls, n=6, P<0.01). Ethanol feeding also reduced creatine kinase release during reperfusion. Inhibition of mitochondrial K(ATP)channels with 5-hydroxydecanoate had no effect on baseline LVDP, LVEDP, or coronary flow but abolished the beneficial effects of alcohol on LV contractile recovery and myocyte necrosis. We conclude that mitochondrial K(ATP)channel activity is required for chronic ethanol-induced protection.


Subject(s)
Ethanol/pharmacology , Ischemic Preconditioning , Mitochondria, Heart/metabolism , Muscle Proteins/physiology , Myocardial Reperfusion Injury/prevention & control , Potassium Channels/physiology , Potassium/physiology , Alcohol Drinking , Animals , Coronary Circulation/drug effects , Creatine Kinase/analysis , Decanoic Acids/pharmacology , Hydroxy Acids/pharmacology , Ion Transport , Isoenzymes/analysis , Myocardial Contraction/drug effects , Necrosis , Rats , Rats, Sprague-Dawley , Ventricular Function, Left/drug effects
4.
Proc Natl Acad Sci U S A ; 96(22): 12784-9, 1999 Oct 26.
Article in English | MEDLINE | ID: mdl-10536000

ABSTRACT

Recent epidemiological studies indicate beneficial effects of moderate ethanol consumption in ischemic heart disease. Most studies, however, focus on the effect of long-term consumption of ethanol. In this study, we determined whether brief exposure to ethanol immediately before ischemia also produces cardioprotection. In addition, because protein kinase C (PKC) has been shown to mediate protection of the heart from ischemia, we determined the role of specific PKC isozymes in ethanol-induced protection. We demonstrated that (i) brief exposure of isolated adult rat cardiac myocytes to 10-50 mM ethanol protected against damage induced by prolonged ischemia; (ii) an isozyme-selective epsilonPKC inhibitor developed in our laboratory inhibited the cardioprotective effect of acute ethanol exposure; (iii) protection of isolated intact adult rat heart also occurred after incubation with 10 mM ethanol 20 min before global ischemia; and (iv) ethanol-induced cardioprotection depended on PKC activation because it was blocked by chelerythrine and GF109203X, two PKC inhibitors. Consumption of 1-2 alcoholic beverages in humans leads to blood alcohol levels of approximately 10 mM. Therefore, our work demonstrates that exposure to physiologically attainable ethanol levels minutes before ischemia provides cardioprotection that is mediated by direct activation of epsilonPKC in the cardiac myocytes. The potential clinical implications of our findings are discussed.


Subject(s)
Adhesins, Bacterial , Carrier Proteins/genetics , Ethanol/therapeutic use , Isoenzymes/metabolism , Myocardial Ischemia/prevention & control , Protein Kinase C/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Carrier Proteins/chemistry , Carrier Proteins/immunology , Dose-Response Relationship, Drug , Enzyme Inhibitors/therapeutic use , In Vitro Techniques , Isoenzymes/antagonists & inhibitors , Male , Molecular Sequence Data , Myocardial Ischemia/enzymology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-epsilon , Rats , Rats, Wistar , Reperfusion Injury/prevention & control
5.
Proc Natl Acad Sci U S A ; 96(22): 12798-803, 1999 Oct 26.
Article in English | MEDLINE | ID: mdl-10536002

ABSTRACT

Brief periods of cardiac ischemia trigger protection from subsequent prolonged ischemia (preconditioning). epsilon Protein kinase C (epsilonPKC) has been suggested to mediate preconditioning. Here, we describe an epsilonPKC-selective agonist octapeptide, psiepsilon receptor for activated C-kinase (psiepsilonRACK), derived from an epsilonPKC sequence homologous to its anchoring protein, epsilonRACK. Introduction of psiepsilonRACK into isolated cardiomyocytes, or its postnatal expression as a transgene in mouse hearts, increased epsilonPKC translocation and caused cardio-protection from ischemia without any deleterious effects. Our data demonstrate that epsilonPKC activation is required for protection from ischemic insult and suggest that small molecules that mimic this epsilonPKC agonist octapeptide provide a powerful therapeutic approach to protect hearts at risk for ischemia.


Subject(s)
Cardiotonic Agents/therapeutic use , Isoenzymes/metabolism , Myocardial Ischemia/prevention & control , Oligopeptides/therapeutic use , Protein Kinase C/metabolism , Receptors, Cell Surface/therapeutic use , Amino Acid Sequence , Animals , Biological Transport , Cardiotonic Agents/chemical synthesis , Cardiotonic Agents/pharmacology , Cell Death/drug effects , In Vitro Techniques , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Protein Kinase C-epsilon , Rats , Rats, Wistar , Receptors for Activated C Kinase
6.
Cardiovasc Res ; 40(2): 352-63, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9893729

ABSTRACT

OBJECTIVE: We sought to determine whether angiotensin II (Ang II) promotes hypertrophy of cardiac directly or via paracrine mechanisms mediated by cardiac fibroblasts. METHODS: We studied neonatal rat cardiac myocytes and fibroblasts in culture as a model system. Paracrine effects of Ang II were identified using conditioned medium and co-culture experiments. RESULTS: Ang II type 1 (AT1) receptors responsible for myocyte growth localized to fibroblasts in radioligand binding, emulsion autoradiography, Western analysis, and immunofluorescence staining experiments. The bulk of AT1 receptor binding in myocyte cultures (1343 +/- 472 sites/cell) was to Ang II receptors on contaminating fibroblasts (9747 +/- 2126 sites/cell). Ang II induced significant paracrine trophic effects on myocytes in conditioned medium (40% increase in protein synthesis over control) and co-culture (4-fold increase over control) experiments. TGF-beta 1 and endothelin-1 were paracrine mediators of hypertrophy in neutralization experiments. CONCLUSIONS: Ang II stimulates cardiac myocyte hypertrophy via paracrine release of TGF-beta 1 and endothelin-1 from cardiac fibroblasts in a neonatal rat cell culture model.


Subject(s)
Angiotensin II/pharmacology , Endothelin-1/metabolism , Myocardium/pathology , Paracrine Communication , Transforming Growth Factor beta/metabolism , Vasoconstrictor Agents/pharmacology , Animals , Animals, Newborn , Blotting, Northern , Blotting, Western , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned/pharmacology , Endothelin-1/analysis , Fibroblasts/drug effects , Fibroblasts/metabolism , Microscopy, Fluorescence , Myocardium/metabolism , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptors, Angiotensin/genetics , Receptors, Angiotensin/metabolism , Transforming Growth Factor beta/analysis
7.
Circ Res ; 81(3): 396-403, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9285642

ABSTRACT

Signaling mediated by the angiotensin (Ang) II and alpha1-adrenergic receptor (alpha1-AR) pathways is important for cardiovascular homeostasis. However, it is unknown whether Ang II has any direct effect on alpha1-AR expression and signaling in cardiac myocytes. In the present study, we determined alpha1-AR subtype mRNA levels by RNase protection; receptor density by competition binding with 5-methylurapidil; and alpha1-AR-mediated c-fos expression by Northern blot analysis. We found that Ang II had no effect on alpha1b- and alpha1d-AR mRNA levels but decreased the alpha1a-AR mRNA level in a time- and dose-dependent manner. The maximal effect occurred at 6 hours with 100 nmol/L Ang II (40.0+/-8.2% reduction, n=4, P<.01). The decrease in alpha1a-AR mRNA level induced by Ang II is mediated by the Ang II AT1 receptor subtype and is associated with decreased stability of alpha1a-AR mRNA. Corresponding to the changes in the alpha1a-AR mRNA level, Ang II (100 nmol/L, 24 hours) reduced the density of high-affinity sites for 5-methylurapidil (alpha1A-AR) by 29% (56.5+/-6.4 versus 79.0+/-11.6 fmol/mg protein, n=4, P<.05). Alpha1-AR-stimulated c-fos induction, which could be blocked by 5-methylurapidil but not by chloroethylclonidine, was attenuated by Ang II preincubation (100 nmol/L, 24 hours). We conclude that there is previously undescribed cross talk between AT1 receptors and alpha1-ARs. Ang II selectively downregulates alpha1a-AR subtype mRNA and its corresponding receptor as well as alpha1a-AR-mediated expression of the immediate-early gene c-fos in cardiac myocytes.


Subject(s)
Angiotensin II/pharmacology , Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Angiotensin/metabolism , Animals , Animals, Newborn , Cells, Cultured , Down-Regulation/drug effects , Genes, fos/drug effects , Myocardium/cytology , Myocardium/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Signal Transduction
8.
J Biol Chem ; 272(3): 1417-20, 1997 Jan 17.
Article in English | MEDLINE | ID: mdl-8999804

ABSTRACT

The protein kinase C (PKC) family consists of 11 isoenzymes. Following activation, each isoenzyme translocates and binds to a specific receptor for activated C kinase (RACK) (Mochly-Rosen, D. (1995) Science 268, 247-251) that provides an anchoring site in close proximity to the isoenzyme's specific substrate. Pancreatic islet cells contain at least six PKC isoenzymes (Knutson, K. L., and Hoenig, M. (1994) Endocrinology 135, 881-886). Although PKC activation enhances insulin release, the specific function of each isoenzyme is unknown. Here we show that following stimulation with glucose, alphaPKC and epsilonPKC translocate to the cell's periphery, while deltaPKC and zetaPKC translocate to perinuclear sites. betaC2-4, a peptide derived from the RACK1-binding site in the C2 domain of betaPKC, inhibits translocation of alphaPKC and reduces insulin response to glucose. Likewise, epsilonV1-2, an epsilonPKC-derived peptide containing the site for its specific RACK, inhibits translocation of epsilonPKC and reduces insulin response to glucose. Inhibition of islet-glucose metabolism with mannoheptulose blocks translocation of both alphaPKC and epsilonPKC and diminishes insulin response to glucose while calcium-free buffer inhibits translocation of alphaPKC but not epsilonPKC and lowers insulin response by 50%. These findings illustrate the unique ability of specific translocation inhibitors to elucidate the isoenzyme-specific functions of PKC in complex signal transduction pathways.


Subject(s)
Islets of Langerhans/enzymology , Isoenzymes/metabolism , Protein Kinase C/metabolism , Animals , Biological Transport , Cells, Cultured , Enzyme Activation , Islets of Langerhans/cytology , Isoenzymes/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Substrate Specificity
9.
J Biol Chem ; 272(49): 30945-51, 1997 Dec 05.
Article in English | MEDLINE | ID: mdl-9388241

ABSTRACT

Protein kinase C activation is thought to protect cardiac tissue from subsequent ischemic injury by a process termed preconditioning. The protein kinase C isozyme that mediates preconditioning has not yet been identified. Using a cell culture model of hypoxic preconditioning, we found that cardiac myocyte viability after 9 h of hypoxia was increased by more than 50% over control. Preconditioning activated protein kinase C isozymes as evidenced by translocation from one cell compartment to another as follows: there was a 2.1-fold increase in epsilon-protein kinase C activation, a 2. 8-fold increase in delta-protein kinase C activation, and no increase in betaI-protein kinase C activation. 4beta-Phorbol 12-myristate 13-acetate mimicked hypoxic preconditioning, increasing myocyte survival after prolonged hypoxia by 34% compared with control. We previously identified an epsilon-protein kinase C-selective antagonist, epsilonV1-2 peptide, that inhibits epsilon-protein kinase C translocation and function in cardiac myocytes (Johnson, J. A., Gray, M. O., Chen, C.-H., and Mochly-Rosen, D. (1996) J. Biol. Chem. 271, 24962-24966). epsilonV1-2 peptide abolished hypoxic preconditioning and phorbol ester-mediated cardiac protection. Therefore, preconditioning can be induced in this culture model, and activation of epsilon-protein kinase C is critical for cardiac myocyte protection.


Subject(s)
Enzyme Inhibitors/pharmacology , Ischemic Preconditioning, Myocardial , Isoenzymes/antagonists & inhibitors , Myocardium/cytology , Peptide Fragments/pharmacology , Protein Kinase C/antagonists & inhibitors , Animals , Biological Transport/drug effects , Cell Death/drug effects , Cell Hypoxia/drug effects , Cells, Cultured , Enzyme Activation/drug effects , HSP70 Heat-Shock Proteins/metabolism , Heart/drug effects , Isoenzymes/physiology , Microscopy, Fluorescence , Protein Kinase C/physiology , Protein Kinase C-epsilon , Rats , Tetradecanoylphorbol Acetate/pharmacology
10.
Circ Res ; 79(6): 1086-99, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8943947

ABSTRACT

We have developed an improved, less disruptive procedure for the transient permeabilization of neonatal cardiac myocytes using saponin. The method allows delivery of peptides to a high percentage of cells in culture without effects on long-term cell viability. Permeation was confirmed microscopically by cellular uptake of a fluorescently labeled peptide and biochemically by uptake of 125I-labeled calmodulin and a 20-kD protein kinase C epsilon fragment into the cells. The intracellular molar concentration of the introduced peptide was approximately 10% of that applied outside. We found no significant effects of permeabilization on spontaneous, phorbol ester-modulated, or norepinephrine-modulated contraction rates. Similarly, the expression of c-fos mRNA (measured 30 minutes after permeabilization) and the incorporation of [-14C]phenylalanine following agonist stimulation (measured 3 days after permeabilization) were not altered by saponin permeabilization. Finally, permeabilization of cells in the presence of a protein kinase C pseudosubstrate peptide, but not a control peptide, inhibited phorbol ester-induced [14C]phenylalanine incorporation into proteins by 80%. Our results demonstrate a methodology for the introduction of peptides into neonatal cardiac myocytes that allows study of their actions without substantial compromises in cell integrity.


Subject(s)
Heart/physiology , Myocardial Contraction/physiology , Peptides/physiology , Protein Kinase C/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/physiology , Protein Kinase C/analysis , Rats , Rats, Sprague-Dawley , Saponins
11.
J Biol Chem ; 271(40): 24962-6, 1996 Oct 04.
Article in English | MEDLINE | ID: mdl-8798776

ABSTRACT

Protein kinase C (PKC) isozymes translocate to unique subcellular sites following activation. We previously suggested that translocation of activated isozymes is required for their function and that in addition to binding to lipids, translocation involves binding of the activated isozymes to specific anchoring proteins (receptors for activated protein kinase C. Using cultured cardiomyocytes we identified inhibitors, the V1 fragment of epsilonPKC (epsilonV1), and an 8-amino acid peptide derived from it that selectively inhibited the translocation of epsilonPKC. Inhibition of epsilonPKC translocation but not inhibition of delta or betaPKC translocation specifically blocked phorbol ester- or norepinephrine-mediated regulation of contraction. These isozyme-selective translocation inhibitors provide novel tools to determine the function of individual PKC isozymes in intact cells.


Subject(s)
Enzyme Inhibitors/pharmacology , Heart/drug effects , Isoenzymes/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Animals , Biological Transport , Cells, Cultured , Isoenzymes/metabolism , Protein Kinase C/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...