Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
2.
J Biol Chem ; 276(22): 19672-7, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11278592

ABSTRACT

Sphingosine 1-phosphate (SPP) binds to members of the endothelial differentiation gene family (EDG) of receptors and leads to diverse signaling events including cell survival, growth, migration and differentiation. However, the mechanisms of how SPP activates these proangiogenic pathways are poorly understood. Here we show that SPP signals through the EDG-1 receptor to the heterotrimeric G protein G(i), leading to activation of the serine/threonine kinase Akt and phosphorylation of the Akt substrate, endothelial nitric-oxide synthase (eNOS). Inhibition of G(i) signaling, and phosphoinositide 3-kinase (PI 3-kinase) activity resulted in a decrease in SPP-induced endothelial cell chemotaxis. SPP also stimulates eNOS phosphorylation and NO release and these effects are also attenuated by inhibition of G(i) signaling, PI 3-kinase, and Akt. However, inhibition of NO production did not influence SPP-induced chemotaxis but effectively blocked the chemotactic actions of vascular endothelial growth factor. Thus, SPP signals through G(i) and PI 3-kinase leading to Akt activation and eNOS phosphorylation.


Subject(s)
Chemotaxis , Endothelium, Vascular/cytology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Lysophospholipids , Nitric Oxide/biosynthesis , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Sphingosine/metabolism , Sphingosine/physiology , Animals , Blotting, Northern , Blotting, Western , Cattle , Cell Movement , Culture Media, Serum-Free/metabolism , Dose-Response Relationship, Drug , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/enzymology , Enzyme Activation , Genes, Dominant , Lung/metabolism , Lymphokines/pharmacology , Neovascularization, Physiologic , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Phosphorylation , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Receptors, Cell Surface/biosynthesis , Signal Transduction , Sphingosine/analogs & derivatives , Time Factors , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Virulence Factors, Bordetella/pharmacology
3.
Nat Med ; 6(9): 1004-10, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10973320

ABSTRACT

Recent studies suggest that statins can function to protect the vasculature in a manner that is independent of their lipid-lowering activity. We show here that statins rapidly activate the protein kinase Akt/PKB in endothelial cells. Accordingly, simvastatin enhanced phosphorylation of the endogenous Akt substrate endothelial nitric oxide synthase (eNOS), inhibited apoptosis and accelerated vascular structure formation in vitro in an Akt-dependent manner. Similar to vascular endothelial growth factor (VEGF) treatment, both simvastatin administration and enhanced Akt signaling in the endothelium promoted angiogenesis in ischemic limbs of normocholesterolemic rabbits. Therefore, activation of Akt represents a mechanism that can account for some of the beneficial side effects of statins, including the promotion of new blood vessel growth.


Subject(s)
Cholesterol/blood , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neovascularization, Physiologic/drug effects , Protein Serine-Threonine Kinases/drug effects , Simvastatin/pharmacology , Animals , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme Activation/drug effects , Hindlimb/blood supply , Male , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Phosphorylation/drug effects , Rabbits , Signal Transduction/drug effects
4.
J Biol Chem ; 275(45): 35291-6, 2000 Nov 10.
Article in English | MEDLINE | ID: mdl-10948187

ABSTRACT

A homeodomain-containing transcription factor Csx/Nkx-2.5 is an important regulator of cardiogenesis in mammals. Three different mutants, Gln170ter (designated A) and Thr178Met (designated B) in the helix 2 of the homeodomain and Gln198ter mutation (designated C) just after homeodomain, have been reported to cause atrial septal defect with atrial ventricular block. We here examined the functions of these three mutants of Csx/Nkx-2.5. The atrial natriuretic peptide (ANP) promoter was activated by wild type Csx/Nkx-2.5 (WT, approximately 8-fold), B ( approximately 2-fold), and C ( approximately 6-fold) but not by A. When A, B, or C was cotransfected into COS-7 cells with the same amount of WT, WT-induced activation of the ANP promoter was attenuated by A and B (A > B), whereas C further enhanced the activation. Immunocytochemical analysis using anti-Myc tag antibody indicated that transfected Myc-tagged WT, B, and C were localized in the nucleus of both COS-7 cells and cardiomyocytes of neonatal rats, whereas A was distributed diffusely in the cytoplasm and nucleus in COS-7 cells. Electrophoretic mobility shift assay showed that Csx/Nkx-2.5-binding sequences were bound strongly by WT and C, weakly by B, but not by A. Immunoprecipitation and GST pull-down assay revealed that WT and all mutants interacted with GATA-4. The synergistic activation of the ANP promoter by WT and GATA-4 was further enhanced by C but was inhibited by A and B. In the cultured cardiomyocytes, overexpression of C but not WT, A, or B, induced apoptosis. These results suggest that although the three mutants induce the same cardiac phenotype, transactivation ability and DNA binding ability are different among the three mutants and that apoptosis may be a cause for C-induced cardiac defect.


Subject(s)
Atrial Natriuretic Factor/genetics , Heart Diseases/congenital , Heart Diseases/etiology , Homeodomain Proteins/genetics , Mutation , Transcription Factors/genetics , Transcription, Genetic , Xenopus Proteins , Animals , Animals, Newborn , Apoptosis , COS Cells , Cell Nucleus/metabolism , Cells, Cultured , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , GATA4 Transcription Factor , Gene Expression Regulation , Genes, Reporter , Glutathione Transferase/metabolism , Heart Septal Defects, Atrial/genetics , Homeobox Protein Nkx-2.5 , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Microscopy, Fluorescence , Myocardium/metabolism , Myocardium/pathology , Nuclear Proteins/metabolism , Phenotype , Plasmids/metabolism , Precipitin Tests , Promoter Regions, Genetic , Protein Binding , Rats , Receptors, Purinergic P1/metabolism , Serum Response Factor , Transcription Factors/metabolism , Transcriptional Activation , Transfection
5.
Biochem Biophys Res Commun ; 272(3): 749-57, 2000 Jun 16.
Article in English | MEDLINE | ID: mdl-10860827

ABSTRACT

Cardiac homeobox gene Csx/Nkx-2.5 is essential for normal heart development and morphogenesis and is the earliest marker for cardiogenesis. To elucidate the regulatory mechanisms of Csx/Nkx-2.5 expression, we have isolated and characterized the upstream regulatory region of human Csx/Nkx-2.5 (CSX1). Transfection of the reporter gene containing a 965-bp CSX1 5' flanking region indicated that this region confers cardiomyocyte-predominant expression of CSX1. Deletion and mutational analyses revealed two positive cis-regulatory elements in this region that are essential for CSX1 expression in cardiomyocytes. Electrophoretic mobility shift assay revealed that nuclear proteins prepared from cardiac myocytes bound to these elements in a sequence-specific manner. The identification of cis-regulatory sequences of the Csx/Nkx-2.5 gene will facilitate further analysis for the upstream regulatory factors that control the expression of Csx/Nkx-2.5 and the process of vertebrate heart development.


Subject(s)
Gene Expression Regulation/genetics , Genes, Homeobox/genetics , Homeodomain Proteins/genetics , Myocardium/metabolism , Promoter Regions, Genetic/genetics , Transcription Factors/genetics , Transcription, Genetic/genetics , Animals , Base Sequence , Binding Sites , COS Cells , Cloning, Molecular , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/metabolism , Homeobox Protein Nkx-2.5 , Humans , Molecular Sequence Data , Mutation/genetics , Myocardium/cytology , Nuclear Proteins/metabolism , Organ Specificity , Response Elements/genetics , Transfection
6.
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
7.
Biochem Biophys Res Commun ; 270(3): 1074-9, 2000 Apr 21.
Article in English | MEDLINE | ID: mdl-10772952

ABSTRACT

A cardiac homeobox-containing gene Csx/Nkx2-5, which is essential for cardiac development, is abundantly expressed in the adult heart as well as in the heart primordia. Targeted disruption of this gene results in embryonic lethality due to abnormal heart morphogenesis. To elucidate the role of Csx/Nkx2-5 in the adult heart, we generated transgenic mice which overexpress human Csx/Nkx2-5. The transgene was expressed abundantly in the heart and the skeletal muscle. mRNA levels of several cardiac genes including natriuretic peptides, CARP, MLC2v, and endogenous Csx/Nkx2-5 were increased in the ventricle of the transgenic mice. Electron microscopic analysis revealed that the ventricular myocardium of the transgenic mice had many secretory granules, which disappeared after administration of vasopressin. These results suggest that Csx/Nkx2-5 regulates many cardiac genes and induces formation of secretory granules in the adult ventricle.


Subject(s)
Atrial Natriuretic Factor/genetics , Cardiac Myosins , Gene Expression Regulation , Homeodomain Proteins/metabolism , Myocardium/metabolism , Transcription Factors/metabolism , Xenopus Proteins , Animals , Heart/embryology , Heart Ventricles , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Humans , Mice , Mice, Transgenic , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Myosin Light Chains/genetics , Natriuretic Peptide, Brain/genetics , Nuclear Proteins/genetics , Protein Isoforms/genetics , Repressor Proteins/genetics , Transcription Factors/genetics
8.
Arterioscler Thromb Vasc Biol ; 20(4): 907-14, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10764653

ABSTRACT

Although smooth muscle cells (SMCs) are critical components of the circulatory system, the regulatory mechanisms of SMC differentiation remain largely unknown. In the present study, we examined the mechanism of SMC differentiation by using Xenopus laevis SM22alpha (XSM22alpha) as an SMC-specific marker. XSM22alpha cDNA contained a 600-bp open reading frame, and the predicted amino acid sequences were highly conserved in evolution. XSM22alpha transcripts were first detected in heart anlage, head mesenchyme, and the dorsal side of the lateral plate mesoderm at the tail-bud stage, possibly representing the precursors of muscle lineage. At the tadpole stage, XSM22alpha transcripts were restricted to the vascular and visceral SMCs. XSM22alpha was strongly induced by basic fibroblast growth factor (FGF) in animal caps. Although expressions of Xenopus cardiac actin were not affected by the expression of a dominant-negative FGF receptor, its injection dramatically suppressed the XSM22alpha expression. These results suggest that XSM22alpha is a useful molecular marker for the SMC lineage in Xenopus and that FGF signaling plays an important role in the induction of XSM22alpha and in the differentiation of SMCs.


Subject(s)
Fibroblast Growth Factor 2/physiology , Gene Expression Regulation, Developmental , Microfilament Proteins , Muscle Proteins/genetics , Xenopus laevis/embryology , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/chemistry , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation, Developmental/drug effects , Molecular Sequence Data , Muscle Proteins/analysis , Muscle Proteins/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution , Xenopus laevis/growth & development
9.
Mol Cell Biol ; 19(10): 7096-105, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10490646

ABSTRACT

Bone morphogenetic proteins (BMPs) have been shown to induce ectopic expression of cardiac transcription factors and beating cardiomyocytes in nonprecardiac mesodermal cells in chicks, suggesting that BMPs are inductive signaling molecules that participate in the development of the heart. However, the precise molecular mechanisms by which BMPs regulate cardiac development are largely unknown. In the present study, we examined the molecular mechanisms by which BMPs induce cardiac differentiation by using the P19CL6 in vitro cardiomyocyte differentiation system, a clonal derivative of P19 embryonic teratocarcinoma cells. We established a permanent P19CL6 cell line, P19CL6noggin, which constitutively overexpresses the BMP antagonist noggin. Although almost all parental P19CL6 cells differentiate into beating cardiomyocytes when treated with 1% dimethyl sulfoxide, P19CL6noggin cells did not differentiate into beating cardiomyocytes nor did they express cardiac transcription factors or contractile protein genes. The failure of differentiation was rescued by overexpression of BMP-2 or addition of BMP protein to the culture media, indicating that BMPs were indispensable for cardiomyocyte differentiation in this system. Overexpression of TAK1, a member of the mitogen-activated protein kinase kinase kinase superfamily which transduces BMP signaling, restored the ability of P19CL6noggin cells to differentiate into cardiomyocytes and concomitantly express cardiac genes, whereas overexpression of the dominant negative form of TAK1 in parental P19CL6 cells inhibited cardiomyocyte differentiation. Overexpression of both cardiac transcription factors Csx/Nkx-2.5 and GATA-4 but not of Csx/Nkx-2.5 or GATA-4 alone also induced differentiation of P19CL6noggin cells into cardiomyocytes. These results suggest that TAK1, Csx/Nkx-2.5, and GATA-4 play a pivotal role in the cardiogenic BMP signaling pathway.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , MAP Kinase Kinase Kinases/metabolism , Myocardium/cytology , Transcription Factors/metabolism , Xenopus Proteins , Bone Morphogenetic Proteins/antagonists & inhibitors , Carrier Proteins , Cell Differentiation , DNA-Binding Proteins/metabolism , GATA4 Transcription Factor , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , MAP Kinase Kinase Kinases/genetics , Models, Biological , Protein Biosynthesis , Proteins/genetics , Recombinant Proteins/metabolism , Transcription Factors/genetics , Tumor Cells, Cultured
12.
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
13.
Ann N Y Acad Sci ; 874: 38-48, 1999 Jun 30.
Article in English | MEDLINE | ID: mdl-10415519

ABSTRACT

Mechanical stretch induced by high blood pressure is an initial factor leading to cardiac hypertrophy. In an in vivo study, an angiotensin II (AngII) type 1 receptor antagonist TCV116 reduced left ventricular (LV) weight, LV wall thickness, transverse myocyte diameter, relative amount of V3 myosin heavy chain, and interstitial fibrosis, while treatment with hydralazine did not. In an in vitro study using cultured cardiomyocytes, mechanical stretch activated second messengers such as mitogen-activated protein (MAP) kinase, followed by increased protein synthesis. Additionally, in the stretch-conditioned medium AngII and endothelin-1 concentrations were increased. Furthermore, the Na+/H+ exchanger activated by mechanical stretch modulated the hypertrophic responses of cardiomyocytes. The pathways leading to MAP kinase activation differed between cell types. In cardiac fibroblasts AngII activated MAP kinase via G beta gamma subunit of Gi, Src, Shc, Grb2, and Ras, whereas Gq and protein kinase C were critical in cardiomyocytes.


Subject(s)
Cardiac Output, Low/metabolism , Cardiomegaly/metabolism , Angiotensin II/physiology , Animals , Cardiac Output, Low/physiopathology , Cardiomegaly/etiology , Cardiomegaly/physiopathology , Endothelin-1/physiology , Hypertension/complications , Myocardium/metabolism , Renin-Angiotensin System/physiology , Signal Transduction/physiology , Sodium-Hydrogen Exchangers/physiology , Stress, Mechanical
14.
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
15.
Jpn Circ J ; 63(5): 425-6, 1999 May.
Article in English | MEDLINE | ID: mdl-10943630

ABSTRACT

Atrial septal defect (ASD) is the most common form of congenital cardiac defect in humans. Recently, point mutations in the cardiac homeobox gene CSX/NKX2-5 have been reported to cause the autosomal dominant form of familial ASD. Notably, all the affected patients exhibit atrioventricular conduction disturbance and some of them died suddenly. The first case of familial ASD with a mutation of the CSX/NKX2-5 gene in a Japanese patient is reported here. Identification of CSX/NKX2-5 mutations in ASD patients would be very important because the existence of such mutations may predict sudden cardiac death.


Subject(s)
Heart Block/genetics , Heart Septal Defects, Atrial/genetics , Homeodomain Proteins/genetics , Transcription Factors/genetics , Heart Block/physiopathology , Heart Septal Defects, Atrial/physiopathology , Homeobox Protein Nkx-2.5 , Humans , Male , Middle Aged , Point Mutation
16.
Jpn Heart J ; 40(6): 803-18, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10737564

ABSTRACT

Left ventricular hypertrophy (LVH) is a secondary adaptation to increased external load. Various qualitative and quantitative changes in myocytes and extracellular components occur during the development of LVH. It has recently been demonstrated that alpha-smooth muscle actin (alpha-SMA)-expressing myofibroblasts appear in the interstitium of the heart subjected to increased workload suggesting that cardiac fibroblasts as well as myocytes alter their phenotype in response to pressure overload. In the present study, to explore the load-induced response and phenotypic modulation of cardiac fibroblasts, the localization of embryonic smooth muscle myosin heavy chain (SMemb) and alpha-SMA in thoracic aorta-constricted rat hearts was investigated by immunohistochemistry, and the morphology of the SMemb-expressing cells was examined by electron microscopy. In addition, to clarify the mechanisms by which SMemb is induced in pressure-overloaded hearts, mRNA expression of SMemb in aorta-constricted rat hearts and in transforming growth factor-beta1 (TGF-beta1)-treated or mechanically-stretched cultured cardiac fibroblasts was investigated. Enhanced staining of SMemb and alpha-SMA was detected in the interstitial spindle-shaped cells in the fibrotic lesions of the pressure-overloaded left ventricles by immunohistochemistry. These cells were demonstrated by electron microscopy to have features specific for activated fibroblasts such as serrated nuclei or prominent rough endoplasmic reticulum. These cells also had characteristic features of myofibroblasts, i.e. irregularly arranged actin filaments and scattered dense bodies. Northern blot analysis revealed increased mRNA levels of SMemb both in aorta-constricted rat hearts and in cultured cardiac fibroblasts stimulated by TGF-beta1 or by mechanical stretch. These results suggest that SMemb may be a molecular marker both for the detection of activated cardiac fibroblasts that may play important roles in the remodeling of pressure-overloaded cardiac interstitium, and for the identification of the regu latory mechanisms that control the phenotypic modulation of cardiac fibroblasts in response to pressure overload.


Subject(s)
Actins/metabolism , Fibroblasts/metabolism , Hypertrophy, Left Ventricular/physiopathology , Muscle, Smooth/embryology , Myocardium/pathology , Myosin Heavy Chains/metabolism , Animals , Fibrosis , Hypertrophy, Left Ventricular/pathology , Immunohistochemistry , Male , Muscle, Smooth/metabolism , Myocardium/metabolism , Pressure , RNA, Messenger/metabolism , Rats , Rats, Wistar
17.
Circulation ; 97(19): 1952-9, 1998 May 19.
Article in English | MEDLINE | ID: mdl-9609089

ABSTRACT

BACKGROUND: Many studies have suggested that the renin-angiotensin system plays an important role in the development of pressure overload-induced cardiac hypertrophy. Moreover, it has been reported that pressure overload-induced cardiac hypertrophy is completely prevented by ACE inhibitors in vivo and that the stored angiotensin II (Ang II) is released from cardiac myocytes in response to mechanical stretch and induces cardiomyocyte hypertrophy through the Ang II type 1 receptor (AT1) in vitro. These results suggest that the AT1-mediated signaling is critical for the development of mechanical stress-induced cardiac hypertrophy. METHODS AND RESULTS: To determine whether AT1-mediated signaling is indispensable for the development of pressure overload-induced cardiac hypertrophy, pressure overload was produced by constricting the abdominal aorta of AT1A knockout (KO) mice. Quantitative reverse transcriptase-polymerase chain reaction revealed that the cardiac AT1 (probably AT1B) mRNA levels in AT1A KO mice were <10% of those of wild-type (WT) mice and were not affected by pressure overload. Chronic treatment with subpressor doses of Ang II increased left ventricular mass in WT mice but not in KO mice. Pressure overload, however, fully induced cardiac hypertrophy in KO as well as WT mice. There were no significant differences between WT and KO mice in expression levels of fetal-type cardiac genes, in the left ventricular wall thickness and systolic function as revealed by the transthoracic echocardiogram, or in the histological changes such as myocyte hypertrophy and fibrosis. CONCLUSIONS: AT1-mediated Ang II signaling is not essential for the development of pressure overload-induced cardiac hypertrophy.


Subject(s)
Angiotensin II/pharmacology , Blood Pressure , Cardiomegaly/physiopathology , Receptors, Angiotensin/deficiency , Animals , Aorta, Abdominal/physiology , Blood Pressure/drug effects , Cardiomegaly/etiology , Cardiomegaly/genetics , Echocardiography , Hypertension/complications , Hypertension/physiopathology , Mice , Mice, Knockout , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Receptor, Angiotensin, Type 1 , Receptors, Angiotensin/biosynthesis , Receptors, Angiotensin/genetics , Receptors, Angiotensin/physiology , Signal Transduction , Transcription, Genetic
18.
Mol Cell Biol ; 18(3): 1580-9, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9488475

ABSTRACT

The Rho family GTP-binding proteins play a critical role in a variety of cytoskeleton-dependent cell functions. In this study, we examined the role of Rho family G proteins in muscle differentiation. Dominant negative forms of Rho family proteins and RhoGDI, a GDP dissociation inhibitor, suppressed transcription of muscle-specific genes, while mutationally activated forms of Rho family proteins strongly activated their transcription. C2C12 cells overexpressing RhoGDI (C2C12RhoGDI cells) did not differentiate into myotubes, and expression levels of myogenin, MRF4, and contractile protein genes but not MyoD and myf5 genes were markedly reduced in C2C12RhoGDI cells. The promoter activity of the myogenin gene was suppressed by dominant negative mutants of Rho family proteins and was reduced in C2C12RhoGDI cells. Expression of myocyte enhancer binding factor 2 (MEF2), which has been reported to be required for the expression of the myogenin gene, was reduced at the mRNA and protein levels in C2C12RhoGDI cells. These results suggest that the Rho family proteins play a critical role in muscle differentiation, possibly by regulating the expression of the myogenin and MEF2 genes.


Subject(s)
GTP-Binding Proteins/metabolism , Guanine Nucleotide Dissociation Inhibitors , Muscles/metabolism , Actins/biosynthesis , Actins/genetics , Animals , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Line , DNA-Binding Proteins/metabolism , GTP-Binding Proteins/biosynthesis , GTP-Binding Proteins/genetics , Humans , MEF2 Transcription Factors , Mice , Muscles/cytology , Myogenic Regulatory Factors/biosynthesis , Myogenin/biosynthesis , Myosin Heavy Chains/biosynthesis , Myosin Heavy Chains/genetics , Transcription Factors/metabolism , Transcription, Genetic , cdc42 GTP-Binding Protein , rac GTP-Binding Proteins , rho Guanine Nucleotide Dissociation Inhibitor alpha , rho-Specific Guanine Nucleotide Dissociation Inhibitors , rhoA GTP-Binding Protein
19.
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
20.
Hypertension ; 31(1): 32-8, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9449387

ABSTRACT

The purpose of the present study was to examine the effects of a long-acting calcium antagonist, amlodipine, on the development of cardiac remodeling. Dihydropyridine calcium antagonists have been used widely for many years in the treatment of hypertension and angina pectoris. It has been reported, however, that a prototype of dihydropyridines, nifedipine, does not reduce mortality of patients with ischemic heart disease, possibly because of reflex stimulation of the sympathetic nervous system. A calcium antagonist, amlodipine, has been reported to have potential benefits by virtue of a gradual onset of action and a long duration of effects. Amlodipine (8 mg/kg per day, once a day) or nifedipine (24 mg/kg per day, three times a day) was administered to spontaneously hypertensive 12-week-old rats for 12 weeks. Left ventricular wall thickness was measured by echocardiography, and relative amounts of myosin heavy chain isoforms were assessed by pyrophosphate gels. Expressions of "fetal type" genes and type 1 collagen gene were examined by Northern blot analysis. Amlodipine and nifedipine both markedly reduced systolic blood pressure. However, the decrease in systolic blood pressure caused by nifedipine continued for no more than 8 hours, whereas the blood pressure-lowering effect of amlodipine continued for more than 16 hours post dose. Amlodipine markedly reduced left ventricular wall thickness, whereas nifedipine only weakly attenuated an increase in the wall thickness. Amlodipine, but not nifedipine, prevented an increase in the relative amount of V3 myosin heavy chain isoform and suppressed an increase in mRNA levels of beta-myosin heavy chain, skeletal alpha-actin, and type 1 collagen. Unlike nifedipine, amlodipine effectively prevented cardiac remodeling secondary to high blood pressure at biochemical levels and morphological levels. These results suggest that a long-acting calcium antagonist is more effective than a short-acting one in preventing organ injury in hypertensive subjects.


Subject(s)
Amlodipine/therapeutic use , Calcium Channel Blockers/therapeutic use , Cardiomegaly/prevention & control , Hypertension/drug therapy , Amlodipine/pharmacology , Animals , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Calcium Channel Blockers/pharmacology , Cardiomegaly/etiology , Collagen/analysis , Collagen/drug effects , Heart/drug effects , Hypertension/complications , Male , Myocardium/chemistry , Myosin Heavy Chains/analysis , Myosin Heavy Chains/drug effects , Nifedipine/pharmacology , Nifedipine/therapeutic use , Rats , Rats, Inbred SHR , Rats, Inbred WKY
SELECTION OF CITATIONS
SEARCH DETAIL
...