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










Publication year range
1.
Nutr Metab Cardiovasc Dis ; 19(10): 707-12, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19346111

ABSTRACT

BACKGROUND AND AIMS: We have previously shown that a maternal low protein (LP) diet during pregnancy results in severe depression of neonatal heart contractility due, in part, to an increase in apoptotic loss of cardiomyocytes. The aim of this study was to examine if maternal LP diet would alter replicative potential of neonatal myocardial cells. METHODS AND RESULTS: We determined the effect of maternal LP and normal diet (90 and 180 g/casein/kg respectively) on relative numbers of mitotic myocardial cells in male offspring at birth and at 7-28 days post-partum. Myocardial cells undergoing mitosis were identified by dual-immunofluorescence of cardiac sections for cardiac muscle myosin and phosphorylated histone 3, whereas cells within the cell cycle were identified by immunoreactivity for Ki67 at 14-28 days post-partum. Neonates from control dams displayed the expected gradual decline in mitotic cells from birth to 28 days post-partum. Hearts from LP offspring had lower numbers of mitotic cells at birth, compared to controls, suggestive of subnormal muscle cell numbers at that stage. When placed in normal diet, LP offspring developed increased myocardial mitosis at 7 days compared to controls, which normalized to control levels at 21-28 days post-partum. An increase in Ki67-positive myocardial cells was also observed in the LP exposed group at 28 days of age. CONCLUSION: Maternal LP diet suppresses myocardial replicative potential and this likely contributes to reduced cell numbers at birth. This suppression is lifted by a protein-replete diet which stimulates post-natal replication of myocardial cells and likely results in a catching-up in cell numbers.


Subject(s)
Diet, Protein-Restricted/adverse effects , Maternal Nutritional Physiological Phenomena , Mitosis/physiology , Myocytes, Cardiac/metabolism , Aging , Animals , Animals, Newborn , Body Weight , Cell Cycle , Cell Proliferation , Female , Heart/growth & development , Male , Myocardium/metabolism , Myocardium/pathology , Organ Size , Pregnancy , Rats
2.
Cell Commun Adhes ; 10(4-6): 293-7, 2003.
Article in English | MEDLINE | ID: mdl-14681031

ABSTRACT

Phosphorylation affects several biological functions of connexin43 (Cx43), although its role on Cx43-mediated inhibition of DNA synthesis is not known. Previous studies showed increased Cx43 phosphorylation on serine in response to growth factor stimulation of cardiomyocytes, mediated by protein kinase C-epsilon (PKCepsilon). Here we report that activation of PKCepsilon is also necessary for stimulation of cardiomyocyte DNA synthesis and mitosis. We have investigated the participation of specific serine residues that are putative PKC targets in producing phosphorylated Cx43 species and also in regulating DNA synthesis in cardiomyocytes. Interference with the PKC signaling system and/or the phosphorylation of specific amino-acids of Cx43 may allow regulation of the mitogenic response.


Subject(s)
Connexin 43/metabolism , Fibroblast Growth Factor 2/pharmacology , Mitosis/drug effects , Phosphorylation/drug effects , Protein Kinase C/metabolism , Animals , Cells, Cultured , Cloning, Molecular , Connexin 43/genetics , DNA Replication , Enzyme Activation/drug effects , HeLa Cells , Humans , Mutation , Myocytes, Cardiac/metabolism , Protein Kinase C-epsilon , Rats , Serine/metabolism
3.
Am J Physiol Heart Circ Physiol ; 280(3): H1039-50, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11179045

ABSTRACT

We generated transgenic (TG) mice overexpressing fibroblast growth factor (FGF)-2 protein (22- to 34-fold) in the heart. Chronic FGF-2 overexpression revealed no significant effect on heart weight-to-body weight ratio or expression of cardiac differentiation markers. There was, however, a significant 20% increase in capillary density. Although there was no change in FGF receptor-1 expression, relative levels of phosphorylated c-Jun NH(2)-terminal kinase and p38 kinase as well as of membrane-associated protein kinase C (PKC)-alpha and total PKC-epsilon were increased in FGF-2-TG mouse hearts. An isolated mouse heart model of ischemia-reperfusion injury was used to assess the potential of increased endogenous FGF-2 for cardioprotection. A significant 34-45% increase in myocyte viability, reflected in a decrease in lactate dehydrogenase released into the perfusate, was observed in FGF-2 overexpressing mice and non-TG mice treated exogenously with FGF-2. In conclusion, FGF-2 overexpression causes augmentation of signal transduction pathways and increased resistance to ischemic injury. Thus, stimulation of endogenous FGF-2 expression offers a potential mechanism to enhance cardioprotection.


Subject(s)
Fibroblast Growth Factor 2/genetics , JNK Mitogen-Activated Protein Kinases , Muscle Fibers, Skeletal/enzymology , Myocardium/cytology , Myocardium/metabolism , Animals , Capillaries/physiology , Cell Survival/physiology , Coronary Circulation/physiology , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Gene Expression/physiology , In Vitro Techniques , MAP Kinase Kinase 4 , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Myocardial Reperfusion Injury/metabolism , Organ Size , Protein Kinase C/metabolism , Transgenes/physiology , p38 Mitogen-Activated Protein Kinases
4.
J Cell Physiol ; 186(3): 457-67, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11169985

ABSTRACT

Fibroblast growth factor-2 (FGF-2) is a mitogen found in CUG-initiated 21-25 kDa ("hi") or AUG-initiated 16-18 kDa ("lo") forms. Previously we demonstrated that "hi"-but not "lo"-FGF-2 caused a distinct nuclear phenotype characterized by apparently condensed chromatin present as separate clumps in the nucleus of cardiac myocytes. In this manuscript we investigated whether these effects were related to apoptosis or mitosis and whether they reflected a direct effect of "hi" FGF-2 on chromatin. Myocytes overexpressing "hi" FGF-2 and presenting the clumped chromatin phenotype: (i) were not labeled above background with antibodies to phosphorylated histones H1 and H3 used as indicators of mitotic chromatin condensation; (ii) did not stain positive for TUNEL; (iii) their nuclear lamina, visualized by anti-laminB immunofluorescence, appeared intact; (iv) neither caspase inhibitors, nor Bcl-2 or "lo" FGF-2 overexpression prevented the manifestation of the compacted nuclear phenotype. Purified recombinant "hi" FGF-2 was more potent than "lo" FGF-2 in promoting the condensation/aggregation of chick erythrocyte chromatin partially reconstituted with histone H1 in vitro. We conclude that the DNA phenotype induced by "hi" FGF-2 in cardiac myocytes likely reflects a direct effect on chromatin structure that does not require the engagement of mitosis or apoptosis. By affecting chromatin compaction "hi" FGF-2 may contribute to the regulation of gene expression.


Subject(s)
Chromatin/physiology , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 2/physiology , Myocardium/cytology , Myocardium/metabolism , Animals , Animals, Newborn , Apoptosis , Base Sequence , Cells, Cultured , Chickens , Chromatin/drug effects , Chromatin/ultrastructure , DNA Primers , Erythrocytes/physiology , Erythrocytes/ultrastructure , Fibroblast Growth Factor 2/genetics , Histones/metabolism , Humans , Kinetics , Mitosis , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/pharmacology , Protein Isoforms/physiology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Transfection
5.
Cell Commun Adhes ; 8(4-6): 253-6, 2001.
Article in English | MEDLINE | ID: mdl-12064598

ABSTRACT

We have used adenoviral vectors to express dominant negative variants of protein kinase C epsilon (PKCepsilon) or mitogen kinase kinase 1 (MKK1) to investigate their involvement in phorbol ester-induced connexin-43 (Cx43) phosphorylation in cardiomyocytes. Stimulation of cardiomyocytes with phorbol 12-myristate 13-acetate (PMA) increased the fraction of the slower migrating (> or = 45 kDa) and more extensively phosphorylated Cx43 species. Expression of dominant negative MKK1 did not prevent the effect of PMA on Cx43 phosphorylation. Selective inhibition of PKCE significantly decreased baseline levels of Cx43 phosphorylation and the PMA-induced accumulation of > or = 45 kDa Cx43. Thus, production of the more extensively phosphorylated species of Cx43 in cardiomyocytes by PMA requires activation of PKCepsilon.


Subject(s)
Connexin 43/metabolism , Isoenzymes/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Myocytes, Cardiac/drug effects , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Cells, Cultured , Genetic Vectors , Isoenzymes/genetics , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase Kinases/genetics , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Phosphorylation , Protein Kinase C/genetics , Protein Kinase C-epsilon , Protein Serine-Threonine Kinases/genetics , Rats
6.
Biochim Biophys Acta ; 1485(1): 1-10, 2000 May 06.
Article in English | MEDLINE | ID: mdl-10802244

ABSTRACT

We examined if elevation in lysophosphatidylethanolamine acyltransferase activity was associated with elevation in phosphatidylethanolamine content during differentiation of P19 teratocarcinoma cells into cardiac myocytes. P19 cells were induced to undergo differentiation into cardiac myocytes by the addition of 1% dimethylsulfoxide to the medium. Immunofluorescence microscopy revealed the presence of striated myosin at 8 days post-dimethylsulfoxide addition confirming differentiation into cardiac cells. The content of phosphatidylethanolamine was increased 2.1-fold (P<0.05) in differentiated cells compared to undifferentiated cells, whereas the content of phosphatidylcholine was reduced 29% (P<0.05). There were no alterations in the pool sizes of other phospholipids, including cardiolipin. The relative abundance of fatty acids in phospholipids of P19 cells was 18:1 > 18:0 > 16:1 = 18:2 > 16:0 = 14:0 > 20:4 and differentiation did not affect the relative amounts of these fatty acids within individual phospholipids. When cells were incubated with [1,3-(3)H]glycerol, radioactivity incorporated into phosphatidylethanolamine was elevated 5.8-fold, whereas radioactivity incorporated into phosphatidylcholine was unaltered. Ethanolaminephosphotransferase, cholinephosphotransferase and membrane CTP:phosphocholine cytidylyltransferase activities were elevated in differentiated cells compared to undifferentiated cells, whereas membrane and cytosolic phospholipase A2 activities were unaltered. Lysophosphatidylethanolamine acyltransferase activities were elevated 2.4-fold (P<0.05). Lysophosphatidylcholine acyltransferase, monolysocardiolipin acyltransferase, acyl-Coenzyme A synthetase and acyl-Coenzyme A hydrolase activities were unaltered in differentiated cells compared to undifferentiated cells. We postulate that during cardiac cell differentiation, the observed elevation in lysophosphatidylethanolamine acyltransferase activity accompanies the elevation in phosphatidylethanolamine mass, possibly to maintain the fatty acyl composition of this phospholipid within the membrane.


Subject(s)
Acyltransferases/metabolism , Myocardium/enzymology , Phospholipids/metabolism , Animals , Carbon Radioisotopes , Cell Differentiation , Cell Membrane/enzymology , Dimethyl Sulfoxide , Fatty Acids/analysis , Glycerol/metabolism , Myocardium/cytology , Myosins/analysis , Oleic Acid/metabolism , Phosphatidylcholines/metabolism , Phosphatidylethanolamines/metabolism , Phospholipids/chemistry , Tritium , Tumor Cells, Cultured
7.
Exp Cell Res ; 256(2): 358-64, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10772808

ABSTRACT

The biosynthesis of phosphatidylethanolamine was examined during differentiation of P19 teratocarcinoma cells into cardiac myocytes. P19 cells were induced to undergo differentiation into cardiac myocytes by the addition of dimethyl sulfoxide to the medium. Immunofluorescence labeling confirmed the expression of striated myosin 10 days postinduction of differentiation. The content of phosphatidylethanolamine increased significantly within the first 2 days of differentiation. [1,3-(3)H]Glycerol incorporation into phosphatidylethanolamine was increased 7.2-fold during differentiation, indicating an elevation in de novo synthesis from 1, 2-diacyl-sn-glycerol. The mechanism for the increase in phosphatidylethanolamine levels during cardiac cell differentiation was a 2.8-fold increase in the activity of ethanolaminephosphotransferase, the 1,2-diacyl-sn-glycerol utilizing reaction of the cytidine 5'-diphosphate-ethanolamine pathway of phosphatidylethanolamine biosynthesis. Incubation of P19 cells with the phosphatidylethanolamine biosynthesis inhibitor 8-(4-chlorophenylthio)-cAMP inhibited the differentiation-induced elevation in phosphatidylethanolamine levels but did not affect the expression of striated myosin. The results suggest that elevation in phosphatidylethanolamine is an early event of P19 cell differentiation into cardiac myocytes, but is not essential for differentiation to proceed.


Subject(s)
Cell Differentiation/physiology , Myocardium/cytology , Phosphatidylethanolamines/biosynthesis , Animals , Desmosomes/metabolism , Mice , Microscopy, Electron , Microscopy, Fluorescence , Myocardium/metabolism , Myocardium/ultrastructure , Myosins/metabolism , Plasmalogens/biosynthesis
8.
Circ Res ; 86(3): 293-301, 2000 Feb 18.
Article in English | MEDLINE | ID: mdl-10679481

ABSTRACT

Gap junctions (GJs), composed of connexins, are intercellular channels ensuring electric and metabolic coupling between cardiomyocytes. We have shown previously that an endogenous mitogenic and cardioprotective protein, fibroblast growth factor-2 (FGF-2), decreases cardiomyocyte GJ permeability by stimulating phosphorylation of connexin-43 (Cx43). Identifying the kinase(s) phosphorylating cardiac Cx43 may thus provide a way of modulating cardiac intercellular communication. Because FGF-2 activates receptors linked to protein kinase C (PKC) and mitogen-activated protein kinase, we first investigated participation of these enzymatic systems in Cx43 phosphorylation. The inhibitor PD98059 blocked activation of mitogen-activated protein kinase, but it did not prevent the FGF-2 effects on GJs. In contrast, the PKC inhibitor chelerythrine blocked the effects of FGF-2 on Cx43 phosphorylation and permeability. Because the epsilon-isoform of PKC localizes to plasma membrane sites, we examined whether it is directly involved in the FGF-2-induced Cx43 phosphorylation. In nonstimulated myocytes, PKCepsilon displayed a discontinuous pattern of localization at intercellular contact sites and partial colocalization with Cx43. Treatment with FGF-2 or phorbol 12-myristate 13-acetate induced a more continuous pattern of PKCepsilon distribution, whereas the anti-Cx43 staining appeared to overlap extensively with that of PKCepsilon. In immunoprecipitation experiments using specific anti-Cx43 antibodies, PKCepsilon but not PKCalpha coprecipitated with Cx43. FGF-2 increased levels of coprecipitated PKCepsilon, suggesting increased association between PKCepsilon and Cx43 on stimulation. Transient gene transfer and overexpression of cDNAs coding for truncated or mutated dominant-negative forms of PKCepsilon decreased cardiomyocyte Cx43 phosphorylation significantly. We conclude that PKC mediates the FGF-2-induced effects on cardiac GJs and that PKCepsilon likely interacts with and phosphorylates cardiac Cx43 at sites of intercellular contact.


Subject(s)
Connexin 43/metabolism , Isoenzymes/physiology , Myocardium/metabolism , Protein Kinase C/physiology , Alkaloids , Animals , Benzophenanthridines , Cells, Cultured , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 2/pharmacology , Gap Junctions/drug effects , Humans , Isoenzymes/antagonists & inhibitors , Mitogen-Activated Protein Kinases/physiology , Myocardium/cytology , Phenanthridines/pharmacology , Phosphorylation/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-epsilon , Rats , Recombinant Proteins/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Tissue Distribution
9.
Cardiovasc Res ; 42(3): 696-705, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10533610

ABSTRACT

OBJECTIVE: The type 1 fibroblast growth factor receptor (FGFR-1) is the only high affinity receptor for fibroblast growth factor-2 (FGF-2) in the rat myocardium, and is essential for normal growth and development of the heart. Levels of FGFR-1 are developmentally regulated, being high in embryonic cardiac myocytes. Also, FGFR-1 exists as both 'long' and 'short' isoforms, and there is a switch from predominant expression of the 'long' isoform in the embryo to the 'short' isoform in the adult heart. Both the decrease in receptor levels and the isoform switch in postnatal cardiac myocytes correlate with a loss of proliferative potential. We investigated whether an increase in either 'long' or 'short' FGFR-1 isoforms could stimulate proliferation in postnatal rat cardiac myocyte cultures. METHODS AND RESULTS: Previously we cloned cDNAs corresponding to 'long' (L) and 'short' (S) FGFR-1 isoforms from embryonic mouse hearts. Hybrid FGFR-1(L) and (S) genes, directed by a myosin light chain-2 promoter and SV40 enhancer sequences, were generated and used to transiently transfect neonatal rat cardiac myocytes. Overexpression of FGFR-1 mRNA and protein was detected by RNA blotting and immunocytochemistry. Ligand-crosslinking confirmed the presence of specific receptors capable of binding FGF-2 on the cell membrane. Overexpression of either FGFR-1(L) or (S) was associated with stimulation of proliferation as assessed by significant increases in bromodeoxyuridine uptake (DNA synthesis) and cell number. To determine whether this response was FGF-2 specific, the level of FGF-2 was assessed in the culture medium of cardiac myocytes overexpressing FGFR-1 isoforms. A three-fold increase was detected in the media of cardiac myocytes overexpressing either FGFR-1(L) or (S) compared to control levels. Neutralization of this FGF-2 with antibodies inhibited the proliferative response. CONCLUSION: Overexpression of either FGFR-1(L) or (S) resulted in an increase in FGF-2-mediated proliferation of postnatal rat cardiac myocytes.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Myocardium/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Fibroblast Growth Factor/genetics , Analysis of Variance , Animals , Animals, Newborn , Cell Count , Cell Division , Cell Membrane/metabolism , Cells, Cultured , Gene Expression , Immunohistochemistry , Microscopy, Fluorescence , Myocardium/cytology , Protein Isoforms/metabolism , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 1 , Receptors, Fibroblast Growth Factor/metabolism
10.
Mol Cell Biochem ; 197(1-2): 187-94, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10485338

ABSTRACT

Cardiac plasma membrane Ca2+/Mg2+ ecto-ATPase (myoglein) requires millimolar concentrations of either Ca2+ or Mg2+ for maximal activity. In this paper, we report its localization by employing an antiserum raised against the purified rat cardiac Ca2+/Mg2+ ATPase. As assessed by Western blot analysis, the antiserum and the purified immunoglobulin were specific for Ca2+/Mg2+ ecto-ATPase; no cross reaction was observed towards other membrane bound enzymes such as cardiac sarcoplasmic reticulum Ca(2+)-pump ATPase or sarcolemmal Ca(2+)-pump ATPase. On the other hand, the cardiac Ca2+/Mg2+ ecto-ATPase was not recognized by antibodies specific for either cardiac sarcoplasmic reticulum Ca(2+)-pump ATPase or plasma membrane Ca(2+)-pump ATPase. Furthermore, the immune serum inhibited the Ca2+/Mg2+ ecto-ATPase activity of the purified enzyme preparation. Immunofluorescence of cardiac tissue sections and neonatal cultured cardiomyocytes with the Ca2+/Mg2+ ecto-ATPase antibodies indicated the localization of Ca2+/Mg2+ ecto-ATPase in association with the plasma membrane of myocytes, in areas of cell-matrix or cell-cell contact. Staining for the Ca2+/Mg2+ ecto-ATPase was not cardiac specific since the antibodies detected the presence of membrane proteins in sections from skeletal muscle, brain, liver and kidney. The results indicate that Ca2+/Mg2+ ecto-ATPase is localized to the plasma membranes of cardiomyocytes as well as other tissues such as brain, liver, kidney and skeletal muscle.


Subject(s)
Adenosine Triphosphatases/metabolism , Myocardium/metabolism , Sarcolemma/enzymology , Adenosine Triphosphatases/immunology , Animals , Cross Reactions , Fluorescent Antibody Technique, Indirect , Immune Sera , Rats , Rats, Sprague-Dawley
11.
Arch Biochem Biophys ; 367(1): 17-25, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10375394

ABSTRACT

There is little information on the molecular events that control the subcellular distribution of protein kinase C during cardiac cell differentiation. We examined protein kinase C activity and the subcellular distribution of representatives of the "classical," "novel," and "atypical" protein kinase C's in P19 murine teratoma cells induced to undergo differentiation into cardiac myocytes by the addition of dimethylsulfoxide to the medium (Grepin et al., Development 124, 2387-2395, 1997). Differentiation was assessed by the presence of striated myosin, a morphological marker for cardiac cells. Addition of dimethyl sulfoxide to the medium resulted in the appearance of striated myosin by 10 days postincubation. Immunolocalization and Western blot studies revealed that a significant proportion of protein kinase Calpha, -epsilon, and -zeta were associated with the particulate fraction in P19 cells prior to differentiation. Differentiation into cardiac cells resulted in a translocation of protein kinase C activity from the particulate fraction to cytosol and localization of most of protein kinase Calpha, -epsilon, and -zeta to the cytoplasmic compartment. The total cellular protein kinase C activity was unaltered during differentiation. The translocation of protein kinase C activity during differentiation of P19 cells into cardiac myocytes was associated with a decrease in the levels of cellular 1, 2-diacyl-sn-glycerol. The cellular levels of phosphatidylserine and phosphatidylinositol did not change during differentiation. Addition of 1,2-dioctanoyl-sn-glycerol, a cell-permeant 1, 2-diacyl-sn-glycerol analog, reversed the differentiation-induced switch in the relative distribution of protein kinase C activity and dramatically increased the association of protein kinase Calpha with the particulate fraction. Addition of 1,2-dioctanoyl-sn-glycerol did not reverse the pattern of distribution for protein kinase Cepsilon or -zeta. The results indicate that protein kinase C activity and protein kinase Calpha, -epsilon and -zeta isoforms are redistributed from the particulate to the cytosolic fraction during differentiation of P19 cells into cardiomyocytes. The mechanism for the redistribution of protein kinase Calpha may be related to the reduction in the cellular 1,2-diacyl-sn-glycerol levels that accompany differentiation.


Subject(s)
Myocardium/cytology , Myocardium/enzymology , Protein Kinase C/metabolism , Animals , Cell Differentiation/drug effects , Cell Lineage , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytosol/drug effects , Cytosol/metabolism , Diglycerides/metabolism , Diglycerides/pharmacology , Dimethyl Sulfoxide/pharmacology , Isoenzymes/metabolism , Mice , Myocardium/metabolism , Myosins/metabolism , Phosphatidylinositols/metabolism , Phosphatidylserines/metabolism , Solubility/drug effects , Teratoma , Tumor Cells, Cultured
12.
Am J Physiol ; 276(3): H826-33, 1999 03.
Article in English | MEDLINE | ID: mdl-10070065

ABSTRACT

Fibroblast growth factor (FGF-2), a mitogenic, angiogenic, and cardioprotective agent, is reported to be released from the postnatal heart by a mechanism of transient remodeling of the sarcolemma during contraction. This release can be increased with adrenergic stimulation. RNA blotting was used to assess whether FGF-2 synthesis in neonatal rat cardiomyocytes might also be regulated by adrenergic stimulation. FGF-2 RNA levels were increased after treatment with norepinephrine for 6 h or with the alpha-adrenergic agonist phenylephrine for 48 h. To assess an effect on transcription, neonatal rat cardiomyocytes were transfected with a hybrid rat FGF-2 promoter/luciferase gene (-1058FGFp.luc) and treated with norepinephrine or phenylephrine for 6 or 48 h, respectively. FGF-2 promoter activity was increased two- to sevenfold in an alpha1-specific manner. Putative phenylephrine-responsive elements (PEREs) were identified at positions -780 and -761 relative to a major transcription initiation site. However, deletion analysis of -1058FGFp.luc showed that the phenylephrine response was independent of the putative PEREs, cell contraction, and Ca2+ influx. In transgenic mice expressing -1058FGFp.luc, a significant three- to sevenfold stimulation of FGF-2 promoter activity was detected in the hearts of two independent lines 6 h after intraperitoneal administration of phenylephrine (50 mg/kg). This increase was still apparent at 24 h but was not detected at 48 h posttreatment. Analysis of FGF-2 mRNA in normal mouse hearts revealed accumulation of the 6.1-kb transcript at 24 h. Control of local FGF-2 synthesis at the transcriptional level through adrenergic stimulation may be important in the response to injury as well as in the maintenance of a healthy myocardium.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Myocardium/metabolism , Promoter Regions, Genetic/physiology , Animals , Base Sequence/genetics , Calcium/metabolism , Gene Expression/drug effects , Male , Mice , Mice, Transgenic/genetics , Mice, Transgenic/metabolism , Molecular Sequence Data , Myocardial Contraction/physiology , Myocardium/cytology , Norepinephrine/pharmacology , Promoter Regions, Genetic/drug effects , RNA/metabolism , Rats , Response Elements/physiology
13.
Biochem Cell Biol ; 77(6): 569-75, 1999.
Article in English | MEDLINE | ID: mdl-10668634

ABSTRACT

Although fibroblast growth factor-2 (FGF-2) plays an important role in cardioprotection and growth, little is known about the signals triggered by it in the adult heart. We therefore examined FGF-2-induced effects on phosphoinositide-specific phospholipase C (PI-PLC) isozymes, which produce second messengers linked to the inotropic and hypertrophic response of the myocardium. FGF-2, administered by retrograde perfusion to the isolated heart, induced an increase in inositol-1,4,5-trisphosphate levels in the cytosol, as well as an increase in total PI-PLC activity associated with sarcolemmal and cytosolic fractions. Furthermore FGF-2 induced a time-dependent elevation in cardiomyocyte membrane-associated PLC gamma1 and PLC beta1 activities, assayed in immunoprecipitated fractions, and moreover, increased the membrane levels of PLC beta1 and PLC beta3. Activation of PLC beta is suggestive of FGF-2-induced cross-talk between FGF-receptor tyrosine kinase and G-protein-coupled signaling in adult cardiomyocytes and underscores the importance of FGF-2 in cardiac physiology.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Isoenzymes/metabolism , Myocardium/enzymology , Type C Phospholipases/metabolism , Animals , Cells, Cultured , Male , Myocardium/cytology , Myocardium/metabolism , Phosphatidylinositol Diacylglycerol-Lyase , Phosphoinositide Phospholipase C , Phospholipase C beta , Phospholipase C gamma , Rats , Rats, Sprague-Dawley , Signal Transduction
14.
J Mol Cell Cardiol ; 30(4): 829-35, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9602432

ABSTRACT

Previous work has demonstrated that drugs which inhibit Na+ entry through voltage-sensitive Na+ channels, or via Na(+)-H+ exchange protect the heart from ischemic reperfusion damage. The purpose of our study was to determine whether these drugs in combination will have an additive protective effect in Langendorff-perfused hearts. During reperfusion following 30 min of ischemia, developed tension and resting tension were 24 +/- 3 and 162 +/- 5%, respectively, of pre-ischemic values in non-treated ischemic hearts. The administration of HOE-642 to inhibit Na+/H+ exchange increased active developed tension (DT) to 58 +/- 2% of pre-ischemic levels and decreased resting tension (RT) to 111 +/- 3% of pre-ischemic levels. The administration of tetrodotoxin (TTX) to block the Na+ channel increased DT to 56 +/- 3% of the pre-ischemic level and reduced the RT to 126 +/- 12% of the pre-ischemic level. Together, HOE-642 and TTX increased recovery of DT to 63 +/- 2% of pre-ischemic levels and improved RT to 116 +/- 4% of pre-ischemic levels after 30 min of reperfusion. All drug treatment protocols significantly lowered the creatine phosphokinase activity measured in the coronary effluent in comparison to that observed in the non-treated hearts. These data demonstrate that inhibition of Na+ entry through either Na(+)-H+ exchange or the Na+ channel protects the heart from ischemic injury, but there is no additional benefit of blocking both routes of Na+ entry simultaneously. This suggests that a threshold level of Na+i may be a critical factor in ischemic cardioprotection.


Subject(s)
Guanidines/pharmacology , Myocardial Ischemia/prevention & control , Myocardial Reperfusion Injury/prevention & control , Sodium Channel Blockers , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium/metabolism , Sulfones/pharmacology , Tetrodotoxin/pharmacology , Animals , Drug Interactions , Heart/physiopathology , Male , Myocardial Ischemia/physiopathology , Rats , Rats, Sprague-Dawley
15.
J Mol Cell Cardiol ; 30(12): 2695-709, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9990540

ABSTRACT

Fibroblast growth factor-2 (FGF-2), administered to the isolated rat heart by perfusion and under constant pressure, is protective against ischemia-reperfusion (I-R). Here we have investigated whether FGF-2 cardioprotection: (a) is dependent on flow modulation; (b) is linked to effects on contractility; (c) is mediated by protein kinase C (PKC); and (d) is linked to PKC and/or mitogen activated protein kinase (MAPK) associated with the sarcolemma. The isolated rat heart was used as a model. Under conditions of constant flow FGF-2 induced significant improvement in recovery of contractile function during I-R. Under constant perfusion pressure, FGF-2 induced a negative inotropic effect (15% decrease in developed pressure). Chelerythrine, a specific PKC inhibitor, prevented both the FGF-2-induced negative inotropic effect before ischemia, and cardioprotection during I-R. FGF-2 induced a chelerythrine-preventable, five-fold increase in sarcolemmal calcium-independent PKC activity. It also increased the association of PKC subtypes -epsilon and -delta with sarcolemmal membranes, detected by Western blotting, as well as, for PKC delta, by immunolocalization. FGF-2 increased the association of PKC epsilon with the membrane fraction of adult cardiomyocyte in culture, confirming that it can affect PKC signaling in cardiomyocytes directly and in a manner similar to its effects in situ. Finally, FGF-2 induced increased active MAPK at sarcolemmal as well as cytosolic sites. Active sarcolemmal MAPK remained elevated when the FGF-2-induced protection was prevented by chelerythrine. In conclusion, we have provided evidence that cardioprotection by FGF-2 is independent of flow modulation. PKC activation mediates both the FGF-2-induced negative inotropic effect before ischemia and the cardioprotective effect assessed during reperfusion, suggesting a cause and effect relationship. Furthermore, FGF-2 cardioprotection is linked to targeting of sarcolemmal sites by calcium-independent PKC.


Subject(s)
Fibroblast Growth Factor 2/antagonists & inhibitors , Myocardial Contraction/drug effects , Phenanthridines/pharmacology , Protein Kinase C/physiology , Sarcolemma/enzymology , Alkaloids , Animals , Benzophenanthridines , Blotting, Western , Calcium/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Cells, Cultured , Cytosol/drug effects , Densitometry , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 2/physiology , Fluorescent Antibody Technique , Male , Membranes/drug effects , Perfusion , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control , Time Factors , Ventricular Function, Left/drug effects
16.
Trends Cardiovasc Med ; 8(4): 180-7, 1998 May.
Article in English | MEDLINE | ID: mdl-21235931

ABSTRACT

Gap junctions (GJ), composed of connexins, are membrane channels that connect and enable communication between neighboring cells and which, in addition to being essential for the coordinated electrical and contractile activity of the heart, may regulate intercellular transmission of signals affecting proliferative growth. Alterations in GJ permeability that have been associated with the regulation of growth can occur acutely through phosphorylation of connexins: fibroblast growth factor-2 (FGF-2) causes decreased coupling and increased phosphorylation of a major cardiomyocyte connexin, connexin43 (Cx43), while stimulating proliferation of cardiomyocytes. On the other hand, transforming growth factor-ß (TGFß) prevents the effects of FGF-2 on Cx43 phosphorylation, as well as canceling the FGF-2-induced proliferation. Parallel to its link with growth regulation, Cx43 phosphorylation emerges as a functionally important end point for delineating cardiac signal transduction pathways elicited by diverse physiologic or pathologic stimuli.

17.
Mol Cell Biochem ; 176(1-2): 89-97, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9406149

ABSTRACT

Basic fibroblast growth factor (FGF-2) plays an important role in myocardial growth and development and in particular cardiac myocyte proliferation. FGF-2 exerts its effects by binding to cell surface receptors (FGFR-1) of the tyrosine kinase family. We have detected the presence of both long and short isoforms of FGFR-1 in embryonic and adult mouse heart. In this report, we have examined the ability of long and short FGFR-1 isoforms to signal a mitogenic response. Assessment of RNA from rat myoblast H9c2 cells by reverse transcriptase-polymerase chain reaction and RNA blotting revealed that they were deficient in transcripts corresponding to long and short FGFR-1 species. Hybrid genes containing the cDNAs coding for long and short FGFR-1 isoforms directed by the myosin light chain-2 promoter and simian virus 40 enhancer sequences, were used to transiently transfect H9c2 cells. Total tyrosine phosphorylation was increased 2.0 and 2.6 fold in H9c2 cells transfected with the long and short FGFR-1 isoforms, respectively, compared to 'control' transfected H9c2 cells. This was accompanied by a 2.1 and 2.0 fold increase in DNA synthesis, as measured by tritiated thymidine incorporation, in H9c2 cells expressing the long and short FGFR-1 isoforms, respectively. To assess effects on proliferation, H9c2 cells were stably transfected with the myosin light chain-2/FGFR-1 cDNA genes. The rate of proliferation was increased 1.6 and 3.1 fold in H9c2 cells stably expressing the long and short FGFR-1 isoforms, respectively, compared to 'control' H9c2 cells. In contrast to non transfected H9c2 cells, treatment of H9c2 cells stably expressing long FGFR-1 with FGF-2 for 24 h resulted in a slight increase (1.3 fold, p < 0.02) in cell number. However, a greater response (1.5 fold, p < 0.0005) was observed with H9c2 cells stably expressing short FGFR-1 after treatment with FGF-2. These results suggest that both long and short FGFR-1 isoforms are capable of signalling a mitogenic response.


Subject(s)
Myocardium/cytology , Receptor Protein-Tyrosine Kinases , Receptors, Fibroblast Growth Factor/physiology , Animals , Cell Culture Techniques , Cell Division/drug effects , Cell Division/genetics , DNA/biosynthesis , DNA, Complementary/genetics , Fibroblast Growth Factor 2/metabolism , Gene Expression/physiology , Gene Transfer Techniques , Phosphorylation , RNA, Messenger/metabolism , Rats , Receptor, Fibroblast Growth Factor, Type 1 , Receptors, Fibroblast Growth Factor/genetics , Tyrosine/chemistry
18.
Mol Cell Biochem ; 176(1-2): 153-61, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9406157

ABSTRACT

Fibroblast growth factor-2 (FGF-2) promotes cardiac myocyte proliferation and has been detected in extracellular as well as cytoplasmic and nuclear compartments. As a first step in examining the participation of intracellular FGF-2 in cardiac myocyte cell cycle we have investigated its localization in proliferative chicken cells during interphase and the various stages of mitosis in culture. We have used a previously characterized and affinity-purified anti-FGF-2 antibody preparation which recognizes the 19-22 kDa variants of chick FGF-2. By immunofluorescence, bright, punctate anti-FGF-2 labelling was observed in 26% of interphase nuclei from myocytes derived from 5 day embryonic heart ventricles; these nuclei were positive for anti-bromodeoxyuridine staining indicating that they are at the S- or G2 phase of the cell cycle. In prophase and metaphase, bright anti-FGF-2 staining was detected in apparent association with chromosomes. During anaphase, however, anti-FGF-2 staining dissociated from chromosomal locations distinctly remaining in strand-like structures in the area of ensuing cleavage furrow formation. In late telophase and cytokinesis, strong staining persisted in the area of the midbody and reappeared in a small fraction of newly formed daughter nuclei. Absorption of the antibody preparation with immobilized FGF-2 eliminated all staining. This dynamic pattern of anti-FGF-2 staining suggests that chick FGF-2 or immunologically related protein(s) not only increase in DNA-synthesizing nuclei but they may play a role in subsequent stages of mitosis and cytokinesis.


Subject(s)
Cell Cycle/physiology , Fibroblast Growth Factor 2/metabolism , Myocardium/cytology , Animals , Antibodies , Cell Division , Chickens , Fibroblast Growth Factor 2/immunology , Fibroblast Growth Factor 2/physiology , Fluorescent Antibody Technique , Heart Ventricles/cytology , Immunoglobulin G , Meiosis/physiology , Mitosis/physiology , Ventricular Function
19.
Exp Cell Res ; 236(1): 127-36, 1997 Oct 10.
Article in English | MEDLINE | ID: mdl-9344592

ABSTRACT

A sequence-specific monoclonal antibody directed against the gap junction protein connexin43 (Cx43) is shown here to be specific for the unphosphorylated form of this protein. In tissues and cultured cells containing different phosphorylated and unphosphorylated forms of Cx43, the antibody detected only the latter as shown by Western blotting of native and alkaline phosphatase-treated samples. Immunohistochemically, this monoclonal antibody did not recognize gap junctions in the vast majority of cultured cardiac myocytes, where nearly all detectable Cx43 is phosphorylated. In contrast, it was able to detect some intracellular Cx43 in tracheal smooth muscle cells and an epithelial cell line (Cl-9 cells), producing patterns of labeling consistent with those seen using a polyclonal antibody that recognizes both phosphorylated and unphosphorylated forms of Cx43. Immunostaining of gap junctions in the cultured cells indicates that both phosphorylated and unphosphorylated Cx43 are present in some assembled gap junctions, suggesting that assembled junctions do not contain exclusively the phosphorylated form of the protein. Annular gap junctions, believed to form as part of the pathway for internalization and degradation of gap junctions, were only occasionally and sparsely labeled by the monoclonal antibody, indicating that complete protein dephosphorylation is not required for uptake and degradation of gap junctions. Furthermore, the ability of this antibody to recognize only unphosphorylated Cx43, and not any of the phosphorylated forms present in the tissues and cell types examined, suggests that a unique phosphorylation site, perhaps present in the epitope recognized by this antibody, must be phosphorylated prior to phosphorylation of Cx43 at other sites.


Subject(s)
Antibodies, Monoclonal , Connexin 43/immunology , Connexin 43/metabolism , Muscle, Smooth/chemistry , Myocardium/chemistry , Animals , Antibody Specificity , Blotting, Western , Cells, Cultured , Connexin 43/analysis , Dogs , Fluorescent Antibody Technique , Gap Junctions/chemistry , Microscopy, Immunoelectron , Muscle Fibers, Skeletal/chemistry , Muscle Fibers, Skeletal/ultrastructure , Muscle, Smooth/cytology , Myocardium/cytology , Phosphorylation , Rabbits , Rats , Rats, Sprague-Dawley , Trachea/cytology
20.
Circ Res ; 79(4): 647-58, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8831488

ABSTRACT

Cardiac gap junction (GJ) channels, composed of connexins, allow electrical and metabolic couplings between cardiomyocytes, properties important for coordinated action of the heart as well as tissue homeostasis and control of growth and differentiation. Fibroblast growth factor-2 (FGF-2) is an endogenous growth-promoting protein, believed to participate in the short- and long-term responses of the heart to injury. We have examined short-term effects of FGF-2 on cardiac myocyte GJ-mediated metabolic coupling, using cultures of neonatal rat cardiomyocytes. FGF-2 decreased coupling between cardiomyocytes assessed by scrape dye loading as well as microinjection and dye transfer within 30 minutes of administration. Genistein blocked the effects of FGF-2. To determine the mechanism, we next assessed the effect of FGF-2 on expression, distribution, and phosphorylation of connexin43 (Cx43), which is a major cardiomyocyte connexin. FGF-2 did not affect Cx43 mRNA or protein accumulation and synthesis, and it did not change Cx43 localization at sites of intercellular contact as assessed by immunostaining with a polyclonal anti-Cx43 antibody raised against a synthetic peptide containing residues 346 to 363 of Cx43. FGF-2, however, decreased staining intensity at sites of intermyocyte contact when a monoclonal anti-Cx43 antibody was used, suggesting a localized masking of epitope(s) recognized by the monoclonal but not the polyclonal antibody. These epitopes appear to reside within residues 261 to 270 of Cx43, as indicated by full quenching of monoclonal antibody staining with synthetic peptides. In addition, FGF-2 induced a more than twofold increase in Cx43 phosphorylation. Phosphoamino acid analysis indicated increased phosphorylation of Cx43 on serine residues. Although tyrosine phosphorylation of Cx43 was not detected in either treated or control cells, a fraction of Cx43 was immunoprecipitated with anti-phosphotyrosine-specific antibodies in FGF-2-treated myocytes, suggesting interaction (and hence coprecipitation) with phosphotyrosine-containing protein(s). In conclusion, we have identified Cx43 and intercellular communication as targets of FGF-2-triggered and tyrosine phosphorylation-dependent signal transduction in cardiac myocytes. It is suggested that phosphorylation of Cx43 on serine induced by FGF-2 contributes to decreased metabolic coupling between cardiomyocytes.


Subject(s)
Cell Communication/drug effects , Connexin 43/metabolism , Fibroblast Growth Factor 2/pharmacology , Myocardium/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Connexin 43/immunology , Epitopes/immunology , Gap Junctions/metabolism , Myocardium/cytology , Phosphorylation/drug effects , Rats , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...