Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Eur J Clin Nutr ; 62(8): 946-52, 2008 Aug.
Article in English | MEDLINE | ID: mdl-17522600

ABSTRACT

OBJECTIVE: To assess the potential impact of a national iron supplementation programme in rural Vietnam. METHODS: The study included questionnaires, focus group discussions of pregnant women and key informant interviews, together with measurements of haemoglobin (Hb) and a stool examination for soil-transmitted helminths. RESULTS: Iron supplementation significantly increased Hb concentration among participants in the second and third trimesters by 0.4 and 0.7 g/dl, respectively (P=0.017 and P<0.001). The risk of anaemia (Hb <10.0 g/dl) was increased significantly by hookworm infestation (P=0.041) and in summer season (P=0.001) and was decreased significantly by taking iron tablets (P=0.041). CONCLUSIONS: The results of this study show that an iron supplementation programme is beneficial as a part of a comprehensive anaemia programme for pregnant women in these communities. These results will be useful for developing improved iron-deficiency anaemia control programs for pregnant women.


Subject(s)
Anemia, Iron-Deficiency/epidemiology , Hemoglobins/analysis , Hookworm Infections/complications , Iron, Dietary/administration & dosage , Pregnancy Complications, Parasitic/blood , Pregnancy Complications/epidemiology , Prenatal Care , Adult , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/etiology , Anemia, Iron-Deficiency/prevention & control , Cross-Sectional Studies , Dietary Supplements , Feces/parasitology , Female , Hookworm Infections/blood , Hookworm Infections/drug therapy , Humans , Iron, Dietary/metabolism , Pregnancy , Pregnancy Complications/blood , Pregnancy Complications/etiology , Pregnancy Complications/prevention & control , Pregnancy Complications, Parasitic/drug therapy , Risk Factors , Rural Health , Vietnam/epidemiology
2.
Biochem Biophys Res Commun ; 289(4): 901-7, 2001 Dec 14.
Article in English | MEDLINE | ID: mdl-11735132

ABSTRACT

Mechanical stress induces various hypertrophic responses including activation of mitogen-activated protein kinases (MAPKs) in cardiac myocytes. Here we examined the role of the small GTP-binding proteins of Rho family and reactive oxygen species (ROS) in stretch-induced activation of p38MAPK in cardiomyocytes. Overexpression of dominant-negative mutants of Rac1 (D.N. Rac1), D.N.RhoA and D.N.Cdc42 suppressed stretch-induced activation of p38MAPK. Overexpression of constitutively active mutants of Rac1 (C.A.Rac1) and C.A.Cdc42 increased the p38MAPK activity in the absence of mechanical stress. Pretreatment with N-acetyl-L-cysteine and N-(2-mercaptopropionyl)-glycine (NAC) suppressed stretch-induced activation of p38MAPK. Mechanical stretch increased intracellular ROS generation, which was abrogated by overexpression of D.N.Rac1 and attenuated by overexpression of D.N.RhoA and D.N.Cdc42. An increase in protein synthesis evoked by mechanical stretch was suppressed by overexpression of D.N.Rac1 and pretreatment with NAC. These results suggest that mechanical stress induces cardiac hypertrophy through the Rac1-ROS-p38MAPK pathway in cardiac myocytes.


Subject(s)
Cardiomegaly/etiology , Cardiomegaly/metabolism , Reactive Oxygen Species/metabolism , Animals , Cardiomegaly/pathology , Cells, Cultured , Enzyme Activation , Gene Expression , Gene Targeting , Mitogen-Activated Protein Kinases/metabolism , Mutation , Myocardium/metabolism , Myocardium/pathology , Rats , Stress, Mechanical , cdc42 GTP-Binding Protein/genetics , p38 Mitogen-Activated Protein Kinases , rac1 GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/genetics
3.
Mol Cell Biochem ; 223(1-2): 35-46, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11681720

ABSTRACT

Growth hormone (GH) has been reported to be useful to treat heart failure. To elucidate whether GH has direct beneficial effects on the heart, we examined effects of GH on oxidative stress-induced apoptosis in cardiac myocytes. TUNEL staining and DNA ladder analysis revealed that hydrogen peroxide (H2O2)-induced apoptosis of cardiomyocytes was significantly suppressed by the pretreatment with GH. GH strongly activated extracellular signal-regulated kinases (ERKs) in cardiac myocytes and the cardioprotective effect of GH was abolished by inhibition of ERKs. Overexpression of dominant negative mutant Ras suppressed GH-stimulated ERK activation. Overexpression of Csk that inactivates Src family tyrosine kinases also inhibited ERK activation evoked by GH. A broad-spectrum inhibitor of protein tyrosine kinases (PTKs), genistein, strongly suppressed GH-induced ERK activation and the cardioprotective effect of GH against apoptotic cell death. GH induced tyrosine phosphorylation of EGF receptor and JAK2 in cardiac myocytes, and an EGF receptor inhibitor tyrphostin AG1478 and a JAK2 inhibitor tyrphostin B42 completely inhibited GH-induced ERK activation. Tyrphostin B42 also suppressed the phosphorylation of EGF receptor stimulated by GH. These findings suggest that GH has a direct protective effect on cardiac myocytes against apoptosis and that the effect of GH is attributed at least in part to the activation of ERKs through Ras and PTKs including JAK2, Src, and EGF receptor tyrosine kinase.


Subject(s)
Apoptosis/physiology , Heart/drug effects , Human Growth Hormone/pharmacology , Myocardium/metabolism , Proto-Oncogene Proteins , Signal Transduction/physiology , Animals , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Genistein/pharmacology , Hydrogen Peroxide/pharmacology , In Situ Nick-End Labeling , Janus Kinase 2 , Mitogen-Activated Protein Kinases/metabolism , Myocardium/cytology , Naphthalenes/pharmacology , Oxidants/pharmacology , Oxidative Stress , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Rats , Rats, Wistar , Tyrphostins/pharmacology , ras Proteins/metabolism , src-Family Kinases/metabolism
4.
J Biol Chem ; 276(30): 28029-36, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11382775

ABSTRACT

Friend of GATA (FOG)-2 is a multi-zinc finger transcriptional corepressor protein that binds specifically to GATA4. Gene targeting studies have demonstrated that FOG-2 is required for normal cardiac morphogenesis, including the development of the coronary vasculature, left ventricular compact zone, and heart valves. To better understand the molecular mechanisms by which FOG-2 regulates these cardiac developmental programs, we screened a mouse day 11 embryo library using a yeast two-hybrid interaction trap with the fifth and sixth zinc fingers of FOG-2 as bait. Using this approach, we isolated clones encoding the orphan nuclear receptors chicken ovalbumin upstream promoter-transcription factor (COUP-TF) 2 and COUP-TF3. COUP-TF2-null embryos die during embryonic development with defective angiogenesis and cardiac defects, a pattern that partly resembles the FOG-2-null phenotype. The interaction between COUP-TF2 and FOG-2 in mammalian cells was confirmed by co-immunoprecipitation of these proteins from transfected COS-7 cells. The sites of binding interaction between COUP-TF2 and FOG-2 were mapped to zinc fingers 5 and 6 and fingers 7 and 8 of FOG-2 and to the carboxyl terminus of the COUP-TF proteins. Binding to COUP-TF2 was specific because FOG-2 did not interact with the ligand-binding domains of retinoid X receptor alpha, glucocorticoid receptor, and peroxisome proliferating antigen receptor gamma, which are related to the COUP-TF proteins. Full-length FOG-2 markedly enhanced transcriptional repression by GAL4-COUP-TF2(117-414), but not by a COUP-TF2 repression domain mutant. Moreover, FOG-2 repressed COUP-TF2dependent synergistic activation of the atrial natriuretic factor promoter by both GATA4 and the FOG-2-independent mutant GATA4-E215K. Taken together, these findings suggest that FOG-2 functions as a corepressor for both GATA and COUP-TF proteins.


Subject(s)
Atrial Natriuretic Factor/genetics , DNA-Binding Proteins/chemistry , Promoter Regions, Genetic , Receptors, Steroid , Transcription Factors/chemistry , 3T3 Cells , Animals , COS Cells , COUP Transcription Factor I , COUP Transcription Factors , Cell Line , DNA, Complementary/metabolism , DNA-Binding Proteins/metabolism , GATA4 Transcription Factor , Gene Library , Glutathione Transferase/metabolism , Ligands , Mice , Mutation , Phenotype , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Glucocorticoid/chemistry , Receptors, Retinoic Acid/chemistry , Recombinant Proteins/metabolism , Retinoid X Receptors , Transcription Factors/metabolism , Transcription, Genetic , Transfection , Two-Hybrid System Techniques , Zinc Fingers
5.
Mol Cell Biochem ; 212(1-2): 177-82, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11108149

ABSTRACT

Angiotensin II (Ang II) evokes a variety of hypertrophic responses such as activation of protein kinases, reprogramming of gene expressions and an increase in protein synthesis in cardiac myocytes. In this study, we examined the role of Rho family small GTP binding proteins (G proteins) in Ang II-induced cardiac hypertrophy. Ang II strongly activated extracellular signal-regulated protein kinases (ERKs) in cardiac myocytes of neonatal rats. Although Ang II-induced activation of ERKs was completely suppressed by an Ang II type 1 receptor antagonist, CV-11974, this activation was not inhibited by the pretreatment with C3 exoenzyme, which abrogates Rho functions. Overexpression of Rho GDP dissociation inhibitor (Rho-GDI), dominant negative mutants of Rac1 (D.N.Rac1), or D.N.Cdc42 had no effects on Ang II-induced activation of transfected ERK2. The promoter activity of skeletal alpha-actin and c-fos genes was increased by Ang II, and the increase was partly inhibited by overexpression of Rho-GDI and the pretreatment with C3 exoenzyme. Ang II increased phenylalanine incorporation into cardiac myocytes by approximately 1.4 fold as compared with control, and this increase was also significantly suppressed by the pretreatment with C3 exoenzyme. These results suggest that the Rho family small G proteins play important roles in Ang II-induced hypertrophic responses in cardiac myocytes.


Subject(s)
Angiotensin II/pharmacology , Heart/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , rho GTP-Binding Proteins/metabolism , Actins/genetics , Animals , Animals, Newborn , Cardiomegaly , Cells, Cultured , Enzyme Activation , Gene Expression Regulation/drug effects , Genes, fos , Heart/drug effects , Mitogen-Activated Protein Kinase 1/genetics , Myocardium/cytology , Promoter Regions, Genetic , Rats , Rats, Wistar , Recombinant Proteins/metabolism , Transfection
6.
J Biol Chem ; 275(44): 34528-33, 2000 Nov 03.
Article in English | MEDLINE | ID: mdl-10931827

ABSTRACT

Apoptosis of cardiac myocytes is one of the causes of heart failure. Here we examine the mechanism by which the activation of beta-adrenergic receptor induces cardiomyocyte apoptosis. Terminal deoxynucleotide transferase-mediated dUTP nick end labeling and DNA ladder analyses revealed that isoproterenol (Iso) induced the apoptosis of cardiac myocytes of neonatal rats through an increase in intracellular Ca(2+) levels. The Iso-induced cardiomyocyte apoptosis was strongly inhibited by the L-type Ca(2+) channel antagonist nifedipine and by the calcineurin inhibitors cyclosporin A and FK506. Iso reduced the phosphorylation levels of the proapoptotic Bcl-2 family protein Bad and induced cytochrome c release from mitochondria to the cytosol through calcineurin activation. Infusion of Iso increased calcineurin activity by approximately 3-fold in the hearts of wild-type mice but not in the hearts of transgenic mice that overexpress dominant negative mutants of calcineurin. Terminal deoxynucleotide transferase-mediated dUTP nick end labeling analysis revealed that infusion of Iso induced apoptosis of cardiac myocytes and that the number of apoptotic cardiomyocytes was significantly less in the hearts of the transgenic mice compared with the wild-type mice. These results suggest that calcineurin plays a critical role in Iso-induced apoptosis of cardiac myocytes, possibly through dephosphorylating Bad.


Subject(s)
Apoptosis/physiology , Calcineurin/physiology , Myocardium/metabolism , Receptors, Adrenergic, beta/physiology , Animals , Calcineurin/genetics , Calcium/metabolism , Cytochrome c Group/metabolism , Cytosol/drug effects , Cytosol/metabolism , In Situ Nick-End Labeling , Isoproterenol/pharmacology , Mice , Mice, Transgenic , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Myocardium/cytology , Phosphorylation , Rats , Rats, Wistar
8.
J Biol Chem ; 275(20): 15239-45, 2000 May 19.
Article in English | MEDLINE | ID: mdl-10809760

ABSTRACT

Endothelin-1 (ET-1) induces cardiac hypertrophy. Because Ca(2+) is a major second messenger of ET-1, the role of Ca(2+) in ET-1-induced hypertrophic responses in cultured cardiac myocytes of neonatal rats was examined. ET-1 activated the promoter of the beta-type myosin heavy chain gene (beta-MHC) (-354 to +34 base pairs) by about 4-fold. This activation was inhibited by chelation of Ca(2+) and the blocking of protein kinase C activity. Similarly, the beta-MHC promoter was activated by Ca(2+) ionophores and a protein kinase C activator. beta-MHC promoter activation induced by ET-1 was suppressed by pretreatment with the calmodulin inhibitor, W7, the Ca(2+)/calmodulin-dependent kinase II (CaMKII) inhibitor, KN62, and the calcineurin inhibitor, cyclosporin A. beta-MHC promoter activation by ET-1 was also attenuated by overexpression of dominant-negative mutants of CaMKII and calcineurin. ET-1 increased the activity of CaMKII and calcineurin in cardiac myocytes. Pretreatment with KN62 and cyclosporin A strongly suppressed ET-1-induced increases in [(3)H]phenylalanine uptake and in cell size. These results suggest that Ca(2+) plays a critical role in ET-1-induced cardiomyocyte hypertrophy by activating CaMKII- and calcineurin-dependent pathways.


Subject(s)
Calcineurin/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cardiomegaly/physiopathology , Endothelin-1/pharmacology , Heart/drug effects , Myocardium/cytology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Animals, Newborn , Calcimycin/pharmacology , Calcineurin/genetics , Calcium/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cells, Cultured , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Heart/physiology , Ionomycin/pharmacology , Kinetics , Models, Cardiovascular , Myosin Heavy Chains/genetics , Promoter Regions, Genetic/drug effects , Rats , Rats, Wistar , Sulfonamides/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Transfection
9.
Circulation ; 102(23): 2873-9, 2000 Dec 05.
Article in English | MEDLINE | ID: mdl-11104747

ABSTRACT

BACKGROUND: Loss of cardiomyocytes by apoptosis is proposed to cause heart failure. Reactive oxygen species induce apoptosis in many types of cells including cardiomyocytes. Because insulin has been reported to have protective effects, we examined whether insulin prevents cardiomyocytes from oxidative stress-induced apoptotic death. METHODS AND RESULTS: Cultured cardiomyocytes of neonatal rats were stimulated by hydrogen peroxide (H(2)O(2)). Apoptosis was evaluated by means of the TUNEL method and DNA laddering. Incubation with 100 micromol/L H(2)O(2) for 24 hours increased the number of TUNEL-positive cardiac myocytes (control, approximately 4% versus H(2)O(2), approximately 23%). Pretreatment with 10(-)(6) mol/L insulin significantly decreased the number of H(2)O(2)-induced TUNEL-positive cardiac myocytes (approximately 12%) and DNA fragmentation induced by H(2)O(2). Pretreatment with a specific phosphatidylinositol 3 kinase (PI3K) inhibitor, wortmannin, and overexpression of dominant negative mutant of PI3K abolished the cytoprotective effect of insulin. Insulin strongly activated both PI3K and the putative downstream effector AKT: Moreover, a proapoptotic protein, BAD:, was significantly phosphorylated and inactivated by insulin through PI3K. CONCLUSIONS: These results suggest that insulin protects cardiomyocytes from oxidative stress-induced apoptosis through the PI3K pathway.


Subject(s)
Apoptosis/drug effects , Enzyme Activation/drug effects , Insulin/pharmacology , Myocardium/cytology , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Protein Serine-Threonine Kinases , Animals , Animals, Newborn , Cells, Cultured , Cytoskeletal Proteins/drug effects , Hydrogen Peroxide/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins c-akt , Rats
10.
Circulation ; 100(20): 2100-7, 1999 Nov 16.
Article in English | MEDLINE | ID: mdl-10562267

ABSTRACT

BACKGROUND: Although anthracyclines, such as daunomycin (DM) and adriamycin, are potent chemotherapeutic agents, they have serious adverse effects, including cardiac toxicity. In the present study, we investigated the molecular mechanisms of DM-induced cardiomyocyte impairment. METHODS AND RESULTS: When cultured cardiac myocytes of neonatal rats were exposed to 1 micromol/L DM for 24 hours, many cells became positive for TUNEL staining, with morphological changes characteristic of apoptosis. Fragmentation of DNA into oligonucleosome-size fragments was recognized by agarose gel electrophoresis in DM-treated myocytes. DM activated 3 members of the mitogen-activated protein kinase (MAPK) family dose-dependently, such as extracellular signal-regulated protein kinases (ERKs), c-Jun NH(2)-terminal kinases, and p38 MAPK in cardiac myocytes. Oxyradical scavengers or Ca(2+) chelators inhibited DM-induced activation of ERKs and p38 MAPK. DM-induced activation of ERKs was also inhibited by overexpression of dominant negative mutants of Ras (D.N.Ras), and the p38 MAPK activation was attenuated by D.N.Rho. The number of DM-induced apoptotic cells was markedly increased when the ERK signaling pathway was selectively blocked by a specific MAPK/ERK kinase inhibitor, PD98059, whereas pretreatment with a specific inhibitor of p38 MAPK, SB203580, significantly reduced the amount of apoptosis. CONCLUSIONS: These results suggest that DM activates MAPKs through reactive oxygen species and Ca(2+) and that the MAPK family plays important roles in DM-induced apoptosis in cardiac myocytes. ERKs protect cardiomyocytes from apoptosis, whereas p38 MAPK is involved in the induction of cardiomyocyte apoptosis.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis , Daunorubicin/therapeutic use , Heart/physiology , Mitogen-Activated Protein Kinases/physiology , Animals , Apoptosis/drug effects , Calcium/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Heart/drug effects , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Wistar , p38 Mitogen-Activated Protein Kinases
11.
FEBS Lett ; 456(1): 103-7, 1999 Jul 30.
Article in English | MEDLINE | ID: mdl-10452539

ABSTRACT

We here examined the role of the interleukin-6 (IL-6) family of cytokines in endothelin-1 (ET-1)-induced hypertrophic responses using cultured cardiac myocytes of neonatal rats. ET-1 induced expression of IL-6 and leukemia inhibitory factor (LIF) genes. ET-1-induced LIF gene expression was abolished by inhibition of protein kinase C activity. ET-1 activated the promoter of atrial natriuretic peptide and beta-type myosin heavy chain genes through the tyrosine kinase pathway and IL-6 receptor gp130. These results suggest that the IL-6 family of cytokines mediates ET-1-induced expression of some fetal genes in cardiac myocytes.


Subject(s)
Endothelin-1/metabolism , Gene Expression Regulation, Developmental , Heart/growth & development , Interleukin-6/genetics , Myocardium/cytology , Animals , Animals, Newborn , Antigens, CD/genetics , Antigens, CD/metabolism , Atrial Natriuretic Factor/drug effects , Atrial Natriuretic Factor/genetics , Benzylamines/pharmacology , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cells, Cultured , Cyclosporine/pharmacology , Cytokine Receptor gp130 , Cytokines/drug effects , Cytokines/genetics , Egtazic Acid/pharmacology , Endothelin-1/pharmacology , Growth Inhibitors/genetics , Growth Inhibitors/pharmacology , Heart/drug effects , Interleukin-6/metabolism , Ionomycin/pharmacology , Ionophores/pharmacology , Leukemia Inhibitory Factor , Lymphokines/genetics , Lymphokines/pharmacology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mutation , Myocardium/metabolism , Myosin Heavy Chains/drug effects , Myosin Heavy Chains/genetics , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Promoter Regions, Genetic , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Wistar , Sulfonamides/pharmacology , Tetradecanoylphorbol Acetate/pharmacology
12.
Circ Res ; 84(4): 458-66, 1999 Mar 05.
Article in English | MEDLINE | ID: mdl-10066681

ABSTRACT

-Mechanical stress induces a variety of hypertrophic responses, such as activation of protein kinases, reprogramming of gene expression, and an increase in protein synthesis. In the present study, to elucidate how mechanical stress induces such events, we examined the role of Rho family small GTP-binding proteins (G proteins) in mechanical stress-induced cardiac hypertrophy. Treatment of neonatal rat cardiomyocytes with the C3 exoenzyme, which abrogates Rho functions, suppressed stretch-induced activation of extracellular signal-regulated protein kinases (ERKs). Overexpression of the Rho GDP dissociation inhibitor (Rho-GDI), dominant-negative mutants of RhoA (DNRhoA), or DNRac1 significantly inhibited stretch-induced activation of transfected ERK2. Overexpression of constitutively active mutants of RhoA slightly activated ERK2 in cardiac myocytes. Overexpression of C-terminal Src kinase, which inhibits functions of the Src family of tyrosine kinases, or overexpression of DNRas had no effect on stretch-induced activation of transfected ERK2. The promoter activity of skeletal alpha-actin and c-fos genes was increased by stretch, and these increases were completely inhibited by either cotransfection of Rho-GDI or pretreatment with C3 exoenzyme. Mechanical stretch increased phenylalanine incorporation into cardiac myocytes by approximately 1.5-fold compared with control, and this increase was also significantly suppressed by pretreatment with C3 exoenzyme. Overexpression of Rho-GDI or DNRhoA did not affect angiotensin II-induced activation of ERK. ERKs were activated by culture media conditioned by stretch of cardiomyocytes without any treatment, but not of cardiomyocytes with pretreatment by C3 exoenzyme. These results suggest that the Rho family of small G proteins plays critical roles in mechanical stress-induced hypertrophic responses.


Subject(s)
Cardiomegaly/etiology , GTP-Binding Proteins/physiology , Guanine Nucleotide Dissociation Inhibitors , Angiotensin II/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cardiomegaly/pathology , Cells, Cultured , Enzyme Activation/physiology , GTP-Binding Proteins/genetics , Mutation/physiology , Myocardium/enzymology , Myocardium/metabolism , Myocardium/pathology , Phenylalanine/metabolism , Rats , Rats, Wistar , Stress, Mechanical , ras Proteins/physiology , rho-Specific Guanine Nucleotide Dissociation Inhibitors , rhoA GTP-Binding Protein , src-Family Kinases/physiology
13.
J Biol Chem ; 274(12): 8231-9, 1999 Mar 19.
Article in English | MEDLINE | ID: mdl-10075728

ABSTRACT

Although the cardiac homeobox gene Csx/Nkx-2.5 is essential for normal heart development, little is known about its regulatory mechanisms. In a search for the downstream target genes of Csx/Nkx-2. 5, we found that the atrial natriuretic peptide (ANP) gene promoter was strongly transactivated by Csx/Nkx-2.5. Deletion and mutational analyses of the ANP promoter revealed that the Csx/Nkx-2.5-binding element (NKE2) located at -240 was required for high level transactivation by Csx/Nkx-2.5. We also found that Csx/Nkx-2.5 and GATA-4 displayed synergistic transcriptional activation of the ANP promoter, and in contrast to previous reports (Durocher, D., Charron, F., Warren, R., Schwartz, R. J., and Nemer, M. (1997) EMBO J. 16, 5687-5696; Lee, Y., Shioi, T., Kasahara, H., Jobe, S. M., Wiese, R. J., Markham, B., and Izumo, S (1998) Mol. Cell. Biol. 18, 3120-3129), this synergism was dependent on binding of Csx/Nkx-2.5 to NKE2, but not on GATA-4-DNA interactions. Although GATA-4 also potentiated the Csx/Nkx-2.5-induced transactivation of the artificial promoter that contains multimerized Csx/Nkx-2.5-binding sites, Csx/Nkx-2.5 reduced the GATA-4-induced transactivation of the GATA-4-dependent promoters. These findings indicate that the cooperative transcriptional regulation mediated by Csx/Nkx-2.5 and GATA-4 is promoter context-dependent and suggest that the complex cis-trans interactions may fine-tune gene expression in cardiac myocytes.


Subject(s)
DNA-Binding Proteins/metabolism , Heart/growth & development , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Binding Sites/genetics , DNA/chemistry , DNA/metabolism , Drug Synergism , GATA4 Transcription Factor , Gene Expression Regulation, Developmental , Homeobox Protein Nkx-2.5 , Humans , Mice , Myocardium/metabolism , Promoter Regions, Genetic , Rats , Sequence Analysis, DNA , Transcriptional Activation
14.
Nihon Rinsho ; 56(10): 2569-75, 1998 Oct.
Article in Japanese | MEDLINE | ID: mdl-9796321

ABSTRACT

Several cardiac serum markers such as cardiac myosin light chain, creatine kinase MB, and cardiac troponins have been shown to be elevated in patients with acute myocardial infarction. These markers are well utilized as clinical indicators for diagnosis and prognosis of myocardial necrosis. Recent studies have strongly indicated that cardiac structural protein troponin T and I are predictive of future cardiac events in patients with acute coronary syndromes including unstable angina pectoris. Immunoassays have also been developed to measure serum levels of cardiac troponins in recent years. These biochemical markers may be useful as a guide for intervention in the near future.


Subject(s)
Coronary Disease/diagnosis , Troponin I/blood , Troponin T/blood , Acute Disease , Aspartate Aminotransferases/blood , Biomarkers/blood , Creatine Kinase/blood , Humans , L-Lactate Dehydrogenase/blood , Myoglobin/blood , Myosin Light Chains/blood , Prognosis , Syndrome
15.
Circ Res ; 82(3): 337-45, 1998 Feb 23.
Article in English | MEDLINE | ID: mdl-9486662

ABSTRACT

Angiotensin II (Ang II) induces hypertrophy of cardiac myocytes and hyperplasia of cardiac fibroblasts. To determine the molecular mechanism by which Ang II displayed different effects on cardiac myocytes and fibroblasts, we examined signal transduction pathways leading to activation of extracellular signal-regulated kinases (ERKs). Ang II-induced ERK activation was abolished by pretreatment with pertussis toxin and by overexpression of the Gbetagamma subunit-binding domain of the beta-adrenergic receptor kinase 1 in cardiac fibroblasts but not in cardiac myocytes. Inhibition of protein kinase C strongly inhibited activation of ERKs by Ang II in cardiac myocytes, whereas inhibitors of tyrosine kinases but not of protein kinase C abolished Ang II-induced ERK activation in cardiac fibroblasts. Overexpression of C-terminal Src kinase (Csk), which inactivates Src family tyrosine kinases, suppressed the activation of transfected ERK in cardiac fibroblasts. Ang II rapidly induced phosphorylation of Shc and association of Shc with Grb2. Cotransfection of the dominant-negative mutant of Ras or Raf-1 kinase abolished Ang II-induced ERK activation in cardiac fibroblasts. Overexpression of Csk or the dominant-negative mutant of Ras had no effects on Ang II-induced ERK activation in cardiac myocytes. These findings suggest that Ang II-evoked signal transduction pathways differ among cell types. In cardiac fibroblasts, Ang II activates ERKs through a pathway including the Gbetagamma subunit of Gi protein, tyrosine kinases including Src family tyrosine kinases, Shc, Grb2, Ras, and Raf-1 kinase, whereas Gq and protein kinase C are important in cardiac myocytes.


Subject(s)
Angiotensin II/pharmacology , GTP-Binding Proteins/physiology , Genes, ras/physiology , Genes, src/physiology , Heart/physiology , Signal Transduction/drug effects , Animals , CSK Tyrosine-Protein Kinase , Calcium-Calmodulin-Dependent Protein Kinases/physiology , DNA/biosynthesis , Fibroblasts/physiology , Multigene Family/physiology , Myocardium/cytology , Protein Kinase C/physiology , Protein-Tyrosine Kinases/physiology , Rats , Rats, Wistar , Receptors, Angiotensin/physiology , src Homology Domains/physiology , src-Family Kinases
16.
Circ Res ; 82(4): 430-7, 1998 Mar 09.
Article in English | MEDLINE | ID: mdl-9506703

ABSTRACT

We have previously reported that stretching of cardiomyocytes activates the phosphorylation cascade of protein kinases, including Raf-1 kinase and mitogen-activated protein (MAP) kinases, followed by an increase in protein synthesis partly through enhanced secretion of angiotensin II and endothelin-1. Membrane proteins, such as ion channels and exchangers, have been postulated to first receive extracellular stimuli and evoke intracellular signals. The present study was performed to determine whether mechanosensitive ion channels and exchangers are involved in stretch-induced hypertrophic responses. Neonatal rat cardiomyocytes cultured on expandable silicone dishes were stretched after pretreatment with a specific inhibitor of stretch-sensitive cation channels (gadolinium and streptomycin), of ATP-sensitive K+ channels (glibenclamide), of hyperpolarization-activated inward channels (CsCl), or of the Na+-H+ exchanger (HOE 694). Pretreatment with gadolinium, streptomycin, glibenclamide, and CsCl did not show any inhibitory effects on MAP kinase activation by mechanical stretch. HOE 694, however, markedly attenuated stretch-induced activation of Raf-1 kinase and MAP kinases by approximately 50% and 60%, respectively, and attenuated stretch-induced increase in phenylalanine incorporation into proteins. In contrast, HOE 694 did not inhibit angiotensin II-and endothelin-1-induced Raf-1 kinase and MAP kinase activation. These results suggest that among many mechanosensitive ion channels and exchangers, the Na+-H+ exchanger plays a critical role in mechanical stress-induced cardiomyocyte hypertrophy.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cardiomegaly/etiology , Ion Channels/physiology , Mechanoreceptors/physiology , Mitogen-Activated Protein Kinase Kinases , Sodium-Hydrogen Exchangers/physiology , Angiotensin II/physiology , Animals , Benzimidazoles/pharmacology , Biphenyl Compounds , Cytoplasm/physiology , Endothelin-1/physiology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Guanidines/pharmacology , Hydrogen-Ion Concentration , MAP Kinase Kinase 1 , Muscle Proteins/biosynthesis , Peptides, Cyclic/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/physiology , Rats , Rats, Wistar , Signal Transduction , Sulfones/pharmacology , Tetrazoles/pharmacology
17.
J Clin Invest ; 100(7): 1813-21, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9312182

ABSTRACT

A growing body of evidence has suggested that oxidative stress causes cardiac injuries during ischemia/reperfusion. Extracellular signal-regulated kinases (ERKs) have been reported to play pivotal roles in many aspects of cell functions and to be activated by oxidative stress in some types of cells. In this study, we examined oxidative stress-evoked signal transduction pathways leading to activation of ERKs in cultured cardiomyocytes of neonatal rats, and determined their role in oxidative stress-induced cardiomyocyte injuries. ERKs were transiently and concentration-dependently activated by hydrogen peroxide (H2O2) in cardiac myocytes. A specific tyrosine kinase inhibitor, genistein, suppressed H2O2-induced ERK activation, while inhibitors of protein kinase A and C or Ca2+ chelators had no effects on the activation. When CSK, a negative regulator of Src family tyrosine kinases, or dominant-negative mutant of Ras or of Raf-1 kinase was overexpressed, activation of transfected ERK2 by H2O2 was abolished. The treatment with H2O2 increased the number of cells stained positive by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling, and induced formation of DNA ladder and activation of CPP32, suggesting that H2O2 induced apoptosis of cardiac myocytes. When H2O2-induced activation of ERKs was selectively inhibited by PD98059, the number of cardiac myocytes which showed apoptotic death was increased. These results suggest that Src family tyrosine kinases, Ras and Raf-1 are critical for ERK activation by hydroxyl radicals and that activation of ERKs may play an important role in protecting cardiac myocytes from apoptotic death following oxidative stress.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Mitogen-Activated Protein Kinases , Myocardium/metabolism , Oxidative Stress , ras Proteins/metabolism , src-Family Kinases/metabolism , Animals , Apoptosis , Calcium/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Free Radicals , Heart/embryology , Hydrogen Peroxide/pharmacology , Myocardium/cytology , Protein Kinase C/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species , Signal Transduction , p38 Mitogen-Activated Protein Kinases
18.
Circulation ; 95(5): 1260-8, 1997 Mar 04.
Article in English | MEDLINE | ID: mdl-9054858

ABSTRACT

BACKGROUND: Although norepinephrine induces cardiac hypertrophy by activating protein kinase A and C through beta- and alpha 1-adrenoceptors, respectively, protein kinase A has been reported to inhibit cell growth in many other cell types. METHODS AND RESULTS: To elucidate the molecular mechanism of norepinephrine-induced hypertrophic responses, we examined the effects of protein kinase A and protein kinase C on the activities of raf-1 kinase and mitogen-activated protein (MAP) kinases and on protein synthesis rates using cultured cardiomyocytes of neonatal rats. Norepinephrine-induced activation of MAP kinases was partially inhibited by either an alpha 1-adrenoceptor blocker (prazosin) or a beta-adrenoceptor blocker (propranolol) and was completely abolished by both blockers. Both a beta-adrenoceptor agonist, isoproterenol, and an alpha 1-adrenoceptor agonist, phenylephrine, increased the activities of raf-1 kinase and MAP kinases and phenylalanine incorporation into proteins. Furthermore, isoproterenol and phenylephrine synergistically activated these kinases and protein synthesis. Similar synergistic activation of MAP kinases was observed when other protein kinase A-activating agents such as forskolin, dibutyryl cAMP, and isobutyl-methylxanthine were used with a protein kinase C-activating agent at the same time. Chelation of extracellular Ca2+ completely abolished isoproterenol- and phenylephrine-evoked MAP kinase activation. CONCLUSIONS: Norepinephrine activates the raf-1 kinase/MAP kinase cascade through both alpha 1- and beta-adrenergic stimulation, and signaling pathways from the two receptors synergistically induce cardiomyocyte hypertrophy.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Heart/drug effects , Myocardium/metabolism , Norepinephrine/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Adrenergic, alpha-1/physiology , Receptors, Adrenergic, beta/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , Adrenergic alpha-1 Receptor Antagonists , Amino Acids/metabolism , Animals , Animals, Newborn , Biological Transport/drug effects , Bucladesine/pharmacology , Calcium/metabolism , Cardiomegaly , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Kinetics , Phenylephrine/pharmacology , Prazosin/pharmacology , Propranolol/pharmacology , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-raf , Rats , Rats, Wistar , Sarcolemma/drug effects , Sarcolemma/metabolism
19.
Heart Vessels ; Suppl 12: 10-4, 1997.
Article in English | MEDLINE | ID: mdl-9476533

ABSTRACT

Csx/Nkx-2.5 is a murine homeobox gene expressed predominantly in cardiocytes and their progenitor cells. The highly lineage-restricted expression pattern of Csx/Nkx-2.5 gene suggests the existence of a positive autoregulatory loop in the transcriptional regulation of Csx/Nkx-2.5. The first intron of CSX1, a human homolog of Csx/Nkx-2.5 gene, had two potential CSX1-binding sequences. Activity of the CSX1 minimal promoter in cultured cardiac myocytes was significantly increased by placing the 3' half of the CSX1 first intron downstream of the reporter gene, suggesting that this region functions as a positive enhancer element. Transient transfection experiments in nonmuscle cells demonstrated that the reporter construct containing the CSX1 minimal promoter and the 3' half of the CSX1 first intron was strongly transactivated by overexpression of CSX1, whereas the CSX1 minimal promoter alone was not. Together these results suggest that the highly lineage-restricted expression of CSX1 is accomplished by autoactivation, which may be mediated by the enhancer element in the first intron.


Subject(s)
Gene Expression Regulation, Developmental , Heart/physiology , Homeodomain Proteins/genetics , Homeostasis , Transcription Factors , Animals , Cells, Cultured , Cloning, Molecular , Enhancer Elements, Genetic/genetics , Exons , Genes, Homeobox , Genes, Reporter/genetics , Genome , Heart/embryology , Homeobox Protein Nkx-2.5 , Humans , Introns , Transcription, Genetic
20.
J Biol Chem ; 271(52): 33592-7, 1996 Dec 27.
Article in English | MEDLINE | ID: mdl-8969227

ABSTRACT

Angiotensin II (AngII) induces cardiac hypertrophy through activating a variety of protein kinases. In this study, to understand how cardiac hypertrophy develops, we examined AngII-evoked signal transduction pathways leading to the activation of extracellular signal-regulated protein kinases (ERKs), which are reportedly critical for the development of cardiac hypertrophy, in cultured cardiac myocytes isolated from neonatal rats. Inhibition of protein kinase C (PKC) with calphostin C or down-regulation of PKC by pretreatment with a phorbol ester for 24 h abolished AngII-induced activation of Raf-1 and ERKs, and addition of a phorbol ester conversely induced a marked increase in the activities of Raf-1 and ERKs. Pretreatment with two chemically and mechanistically dissimilar tyrosine kinase inhibitors, genistein and tyrphostin, did not attenuate AngII-induced activation of ERKs. In contrast, genistein strongly blocked insulin-induced ERK activation in cardiac myocytes. Although pretreatment with manumycin, a Ras farnesyltransferase inhibitor, or overexpression of a dominant-negative mutant of Ras inhibited insulin-induced ERK activation, neither affected AngII-induced activation of ERKs. Overexpression of a dominant-negative mutant of Raf-1 completely suppressed ERK2 activation by AngII, endothelin-1, and insulin. These results suggest that PKC and Raf-1, but not tyrosine kinases or Ras, are critical for AngII-induced activation of ERKs in cardiac myocytes.


Subject(s)
Angiotensin II/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Myocardium/enzymology , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , ras Proteins/metabolism , Animals , Endothelin-1/pharmacology , Enzyme Activation , Genistein , Insulin/pharmacology , Isoflavones/pharmacology , Mitogen-Activated Protein Kinase 1 , Polyenes/pharmacology , Polyunsaturated Alkamides , Proto-Oncogene Proteins c-raf , Rats , Rats, Wistar , Tetradecanoylphorbol Acetate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...