Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
PLoS One ; 7(4): e35632, 2012.
Article in English | MEDLINE | ID: mdl-22558183

ABSTRACT

BACKGROUND: Pressure overload and prolonged angiotensin II (Ang II) infusion elicit cardiac hypertrophy in Ang II receptor 1 (AT(1)) null mouse, whereas Ang II receptor 2 (AT(2)) gene deletion abolishes the hypertrophic response. The roles and signals of the cardiac AT(2) receptor still remain unsettled. Promyelocytic leukemia zinc finger protein (PLZF) was shown to bind to the AT(2) receptor and transmit the hypertrophic signal. Using PLZF knockout mice we directed our studies on the function of PLZF concerning the cardiac specific transcription factor GATA4, and GATA4 targets. METHODOLOGY AND PRINCIPAL FINDINGS: PLZF knockout and age-matched wild-type (WT) mice were treated with Ang II, infused at a rate of 4.2 ng·kg(-1)·min(-1) for 3 weeks. Ang II elevated systolic blood pressure to comparable levels in PLZF knockout and WT mice (140 mmHg). WT mice developed prominent cardiac hypertrophy and fibrosis after Ang II infusion. In contrast, there was no obvious cardiac hypertrophy or fibrosis in PLZF knockout mice. An AT(2) receptor blocker given to Ang II-infused wild type mice prevented hypertrophy, verifying the role of AT(2) receptor for cardiac hypertrophy. Chromatin immunoprecipitation and electrophoretic mobility shift assay showed that PLZF bound to the GATA4 gene regulatory region. A Luciferase assay verified that PLZF up-regulated GATA4 gene expression and the absence of PLZF expression in vivo produced a corresponding repression of GATA4 protein. CONCLUSIONS: PLZF is an important AT(2) receptor binding protein in mediating Ang II induced cardiac hypertrophy through an AT(2) receptor-dependent signal pathway. The angiotensin II-AT(2)-PLZF-GATA4 signal may further augment Ang II induced pathological effects on cardiomyocytes.


Subject(s)
Angiotensin II/adverse effects , Cardiomegaly/metabolism , Fibrosis/metabolism , GATA4 Transcription Factor/genetics , Kruppel-Like Transcription Factors/deficiency , Receptor, Angiotensin, Type 2/metabolism , Angiotensin II/administration & dosage , Angiotensin II Type 2 Receptor Blockers/administration & dosage , Animals , Binding Sites , Blood Pressure/drug effects , Cardiomegaly/chemically induced , Cardiomegaly/complications , Cardiomegaly/physiopathology , Fibrosis/chemically induced , Fibrosis/complications , Fibrosis/physiopathology , GATA4 Transcription Factor/metabolism , Gene Expression Regulation/drug effects , Heart/drug effects , Heart/physiopathology , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Promyelocytic Leukemia Zinc Finger Protein , Protein Binding , Receptor, Angiotensin, Type 2/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription, Genetic/drug effects
2.
Hypertension ; 55(1): 161-5, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19901155

ABSTRACT

Vascular smooth muscle cell hypertrophy, proliferation, or migration occurs in hypertension, atherosclerosis, and restenosis after angioplasty, leading to pathophysiological vascular remodeling. Angiotensin II and platelet-derived growth factor are well-known participants of vascular remodeling and activate a myriad of downstream protein kinases, including p21-activated protein kinase (PAK1). PAK1, an effector kinase of small GTPases, phosphorylates several substrates to regulate cytoskeletal reorganization. However, the exact role of PAK1 activation in vascular remodeling remains to be elucidated. Here, we have hypothesized that PAK1 is a critical target of intervention for the prevention of vascular remodeling. Adenoviral expression of dominant-negative PAK1 inhibited angiotensin II-stimulated vascular smooth muscle cell migration. It also inhibited vascular smooth muscle cell proliferation induced by platelet-derived growth factor. PAK1 was activated in neointima of the carotid artery after balloon injury in the rat. Moreover, marked inhibition of the neointima hyperplasia was observed in a dominant-negative PAK1 adenovirus-treated carotid artery after the balloon injury. Taken together, these results suggest that PAK1 is involved in both angiotensin II and platelet-derived growth factor-mediated vascular smooth muscle cell remodeling, and inactivation of PAK1 in vivo could be effective in preventing pathophysiological vascular remodeling.


Subject(s)
Carotid Artery Injuries/therapy , Carotid Artery, Common/pathology , Myocytes, Smooth Muscle/metabolism , p21-Activated Kinases/metabolism , Angioplasty, Balloon/adverse effects , Angiotensin II/pharmacology , Animals , Becaplermin , Carotid Artery Injuries/etiology , Carotid Artery Injuries/genetics , Carotid Artery, Common/metabolism , Carotid Artery, Common/physiopathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Genetic Therapy/methods , Immunoblotting , Immunohistochemistry , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-sis , Rats , Rats, Sprague-Dawley , Transfection , Tunica Intima/metabolism , Tunica Intima/pathology , p21-Activated Kinases/genetics
3.
Arterioscler Thromb Vasc Biol ; 29(2): 217-24, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19095998

ABSTRACT

BACKGROUND: Although, endothelial nitric oxide (NO) synthase (eNOS) is believed to antagonize vascular remodeling induced by the angiotensin II (AngII) type-1 receptor, the exact signaling mechanism remains unclear. METHODS AND RESULTS: By expressing eNOS to vascular smooth muscle cells (VSMCs) via adenovirus, we investigated a signal transduction mechanism of the eNOS gene transfer in preventing vascular remodeling induced by AngII. We found marked inhibition of AngII-induced Rho/Rho-kinase activation and subsequent VSMC migration by eNOS gene transfer whereas G(q)-dependent transactivation of the epidermal growth factor receptor by AngII remains intact. This could be explained by the specific inhibition of G(12/13) activation by eNOS-mediated G(12/13) phosphorylation. CONCLUSIONS: The eNOS/NO cascade specifically targets the Rho/Rho-kinase system via inhibition of G(12/13) to prevent vascular migration induced by AngII, representing a novel signal cross-talk in cardiovascular protection by NO.


Subject(s)
Cell Movement , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Nitric Oxide Synthase Type III/metabolism , Receptor, Angiotensin, Type 1/metabolism , rho-Associated Kinases/metabolism , Adenoviridae/genetics , Angiotensin II/metabolism , Animals , Cattle , Cells, Cultured , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Endothelial Cells/enzymology , ErbB Receptors/metabolism , Genetic Vectors , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Phosphorylation , Protein Phosphatase 1/metabolism , Rats , Signal Transduction , Time Factors , Transduction, Genetic
4.
Endocrinology ; 149(7): 3569-75, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18356277

ABSTRACT

The angiotensin II (AngII) type 1 receptor (AT(1)) plays a critical role in hypertrophy of vascular smooth muscle cells (VSMCs). Although it is well known that G(q) is the major G protein activated by the AT(1) receptor, the requirement of G(q) for AngII-induced VSMC hypertrophy remains unclear. By using cultured VSMCs, this study examined the requirement of G(q) for the epidermal growth factor receptor (EGFR) pathway, the Rho-kinase (ROCK) pathway, and subsequent hypertrophy. AngII-induced intracellular Ca(2+) elevation was completely inhibited by a pharmacological G(q) inhibitor as well as by adenovirus encoding a G(q) inhibitory minigene. AngII (100nm)-induced EGFR transactivation was almost completely inhibited by these inhibitors, whereas these inhibitors only partially inhibited AngII (100nm)-induced phosphorylation of a ROCK substrate, myosin phosphatase target subunit-1. Stimulation of VSMCs with AngII resulted in an increase of cellular protein and cell volume but not in cell number. The G(q) inhibitors completely blocked these hypertrophic responses, whereas a G protein-independent AT(1) agonist did not stimulate these hypertrophic responses. In conclusion, G(q) appears to play a major role in the EGFR pathway, leading to vascular hypertrophy induced by AngII. Vascular G(q) seems to be a critical target of intervention against cardiovascular diseases associated with the enhanced renin-angiotensin system.


Subject(s)
Angiotensin II/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Myocytes, Smooth Muscle/drug effects , Signal Transduction/drug effects , Adenoviridae/genetics , Animals , Calcium/metabolism , Cell Enlargement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cyclic AMP/metabolism , ErbB Receptors/metabolism , ErbB Receptors/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Hypertrophy , Immunoblotting , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Peptide Fragments/genetics , Peptide Fragments/physiology , Phosphorylation/drug effects , Protein Phosphatase 1/metabolism , Rats , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 1/physiology , rho-Associated Kinases/metabolism
5.
Hypertension ; 51(2): 232-8, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18180404

ABSTRACT

We have shown previously that activation of protein kinase C-delta (PKC delta) is required for angiotensin II (Ang II)-induced migration of vascular smooth muscle cells (VSMCs). Here, we have hypothesized that PKC delta phosphorylation at Tyr(311) plays a critical role in VSMC hypertrophy induced by Ang II. Immunoblotting was used to monitor PKC delta phosphorylation at Tyr(311), and cell size and protein measurements were used to detect hypertrophy in VSMCs. PKC delta was rapidly (0.5 to 10.0 minutes) phosphorylated at Tyr(311) by Ang II. This phosphorylation was markedly blocked by an Src family kinase inhibitor and dominant-negative Src but not by an epidermal growth factor receptor kinase inhibitor. Ang II-induced Akt phosphorylation and hypertrophic responses were significantly enhanced in VSMCs expressing PKC delta wild-type compared with VSMCs expressing control vector, whereas the enhancements were markedly diminished in VSMCs expressing a PKC delta Y311F mutant. Also, these responses were significantly inhibited in VSMCs expressing kinase-inactive PKC delta K376A compared with VSMCs expressing control vector. From these data, we conclude that not only PKC delta kinase activation but also the Src-dependent Tyr(311) phosphorylation contributes to Akt activation and subsequent VSMC hypertrophy induced by Ang II, thus signifying a novel molecular mechanism for enhancement of cardiovascular diseases induced by Ang II.


Subject(s)
Angiotensin II/pharmacology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , Amino Acid Sequence , Animals , Aorta , Cells, Cultured , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Hypertrophy , Immunoblotting , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Phosphorylation , Rats , Receptor, Angiotensin, Type 1/metabolism , Tyrosine , src-Family Kinases/metabolism
6.
Clin Sci (Lond) ; 112(8): 417-28, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17346243

ABSTRACT

The intracellular signal transduction of AngII (angiotensin II) has been implicated in cardiovascular diseases, such as hypertension, atherosclerosis and restenosis after injury. AT(1) receptor (AngII type-1 receptor), a G-protein-coupled receptor, mediates most of the physiological and pathophysiological actions of AngII, and this receptor is predominantly expressed in cardiovascular cells, such as VSMCs (vascular smooth muscle cells). AngII activates various signalling molecules, including G-protein-derived second messengers, protein kinases and small G-proteins (Ras, Rho, Rac etc), through the AT(1) receptor leading to vascular remodelling. Growth factor receptors, such as EGFR (epidermal growth factor receptor), have been demonstrated to be 'trans'-activated by the AT(1) receptor in VSMCs to mediate growth and migration. Rho and its effector Rho-kinase/ROCK are also implicated in the pathological cellular actions of AngII in VSMCs. Less is known about the endothelial AngII signalling; however, recent studies suggest the endothelial AngII signalling positively, as well as negatively, regulates the NO (nitric oxide) signalling pathway and, thereby, modulates endothelial dysfunction. Moreover, selective AT(1)-receptor-interacting proteins have recently been identified that potentially regulate AngII signal transduction and their pathogenic functions in the target organs. In this review, we focus our discussion on the recent findings and concepts that suggest the existence of the above-mentioned novel signalling mechanisms whereby AngII mediates the formation of cardiovascular diseases.


Subject(s)
Angiotensin II/metabolism , Cardiovascular Diseases/metabolism , Endothelium, Vascular/metabolism , Muscle, Smooth, Vascular/metabolism , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/physiology , GTP-Binding Proteins/metabolism , Humans , Receptors, Growth Factor/metabolism , Renin-Angiotensin System/physiology
7.
Endocrinology ; 147(12): 5914-20, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16980435

ABSTRACT

Enhanced angiotensin II (AngII) action has been implicated in endothelial dysfunction that is characterized as decreased nitric oxide availability. Although endothelial cells have been reported to express AngII type 1 (AT1) receptors, the exact role of AT1 in regulating endothelial NO synthase (eNOS) activity remains unclear. We investigated the possible regulation of eNOS through AT1 in bovine aortic endothelial cells (BAECs) and its functional significance in rat aortic vascular smooth muscle cells (VSMCs). In BAECs infected with adenovirus encoding AT1 and in VSMCs infected with adenovirus encoding eNOS, AngII rapidly stimulated phosphorylation of eNOS at Ser1179. This was accompanied with increased cGMP production. These effects were blocked by an AT1 antagonist. The cGMP production was abolished by a NOS inhibitor as well. To explore the importance of eNOS phosphorylation, VSMCs were also infected with adenovirus encoding S1179A-eNOS. AngII did not stimulate cGMP production in VSMCs expressing S1179A. However, S1179A was able to enhance basal NO production as confirmed with cGMP production and enhanced vasodilator-stimulated phosphoprotein phosphorylation. Interestingly, S1179A prevented the hypertrophic response similar to wild type in VSMCs. From these data, we conclude that the AngII/AT1 system positively couples to eNOS via Ser1179 phosphorylation in ECs and VSMCs if eNOS and AT1 coexist. However, basal level NO production may be sufficient for prevention of AngII-induced hypertrophy by eNOS expression. These data demonstrate a novel molecular mechanism of eNOS regulation and function and thus provide useful information for eNOS gene therapy under endothelial dysfunction.


Subject(s)
Nitric Oxide Synthase Type III/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 1/physiology , Angiotensin II/pharmacology , Animals , Aorta/cytology , Cattle , Cells, Cultured , Endothelium, Vascular/metabolism , Enzyme Activation , Gene Transfer Techniques , Hypertrophy/genetics , Phosphorylation/drug effects , Rats , Transfection
9.
Arterioscler Thromb Vasc Biol ; 26(9): e133-7, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16840716

ABSTRACT

BACKGROUND: Angiotensin II (Ang II) promotes growth of vascular smooth muscle cells (VSMCs) via epidermal growth factor (EGF) receptor (EGFR) transactivation mediated through a metalloprotease-dependent shedding of heparin-binding EGF-like growth factor (HB-EGF). However, the identity of the metalloprotease responsible for this process remains unknown. METHODS AND RESULTS: To identify the metalloprotease required for Ang II-induced EGFR transactivation, primary cultured aortic VSMCs were infected with retrovirus encoding dominant negative (dn) mutant of ADAM10 or ADAM17. EGFR transactivation induced by Ang II was inhibited in VSMCs infected with dnADAM17 retrovirus but not with dnADAM10 retrovirus. However, Ang II comparably stimulated intracellular Ca2+ elevation and JAK2 tyrosine phosphorylation in these VSMCs. In addition, dnADAM17 inhibited HB-EGF shedding induced by Ang II in A10 VSMCs expressing the AT1 receptor. Moreover, Ang II enhanced protein synthesis and cell volume in VSMCs infected with control retrovirus, but not in VSMCs infected with dnADAM17 retrovirus. CONCLUSIONS: ADAM17 activated by the AT1 receptor is responsible for EGFR transactivation and subsequent protein synthesis in VSMCs. These findings demonstrate a previously missing molecular mechanism by which Ang II promotes vascular remodeling.


Subject(s)
ADAM Proteins/metabolism , Angiotensin II/pharmacology , ErbB Receptors/genetics , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Transcriptional Activation , ADAM Proteins/genetics , ADAM17 Protein , Animals , Cells, Cultured , Genes, Dominant , Hypertrophy , Mutation , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Rats , Receptor, Angiotensin, Type 1/metabolism , Transfection
10.
J Biol Chem ; 281(31): 21878-21883, 2006 Aug 04.
Article in English | MEDLINE | ID: mdl-16762922

ABSTRACT

SHP-1 and SHP-2 are two Src homology 2 domain-containing tyrosine phosphatases with major pathological implications in cell growth regulating signaling. They share significant overall sequence identity, but their biological functions are often opposite. SHP-1 is generally considered as a negative signal transducer and SHP-2 as a positive one. However, the precise role of each enzyme in shared signaling pathways is not well defined. In this study, we investigated the interaction of these two enzymes in a single cell system by knocking down their expressions with small interfering RNAs and analyzing the effects on epidermal growth factor signaling. Interestingly, knockdown of either SHP-1 or SHP-2 caused significant reduction in the activation of ERK1/2 but not Akt. Furthermore, SHP-1, SHP-2, and Gab1 formed a signaling complex, and SHP-1 and SHP-2 interact with each other. The interaction of SHP-1 with Gab1 is mediated by SHP-2 because it was abrogated by knockdown of SHP-2, and SHP-2, but not SHP-1, binds directly to tyrosine-phosphorylated Gab1. Together, the data revealed that both SHP-1 and SHP-2 have a positive role in epidermal growth factor-induced ERK1/2 activation and that they act cooperatively rather than antagonistically. The interaction of SHP-1 and SHP-2 may be responsible for previously unexpected novel regulatory mechanism of cell signaling by tyrosine phosphatases.


Subject(s)
Epidermal Growth Factor/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Protein Tyrosine Phosphatases/physiology , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Caco-2 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/genetics , RNA, Small Interfering/pharmacology
11.
Curr Pharm Biotechnol ; 7(2): 81-6, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16724941

ABSTRACT

Reactive oxygen species (ROS) are proposed to induce cardiovascular diseases, such as atherosclerosis and hypertension, through several mechanisms. One such mechanism involves ROS acting as intracellular second messengers, which lead to induction of unique signal transductions. Angiotensin II (AngII), a potent cardiovascular pathogen, stimulates ROS production through vascular NADPH oxidases. The ROS production induced by AngII activates downstream ROS-sensitive kinases that are critical in mediating cardiovascular remodeling. Recent advances in gene transfer/knockout techniques have lead to numerous in vitro and in vivo studies that identify the potential components and mechanisms of ROS signal transduction by AngII which promote cardiovascular remodeling. In this review, we will focus our discussion on the signal transduction research elucidating ROS production and function induced by AngII using currently available molecular biotechnologies.


Subject(s)
Angiotensin II/physiology , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Animals , Humans , Reactive Oxygen Species/pharmacology
12.
Curr Vasc Pharmacol ; 4(1): 67-78, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16472178

ABSTRACT

Accumulating evidence strongly implicates angiotensin II (AngII) intracellular signaling in mediating cardiovascular diseases such as hypertension, atherosclerosis and restenosis after vascular injury. In vascular smooth muscle cells (VSMCs), through its G-protein-coupled AngII Type 1 receptor (AT(1)), AngII activates various intracellular protein kinases, such as receptor or non-receptor tyrosine kinases, which includes epidermal growth factor receptor (EGFR), platelet-derived growth factor receptor (PDGFR), c-Src, PYK2, FAK, JAK2. In addition, AngII activates serine/threonine kinases such as mitogen-activated protein kinase (MAPK) family, p70 S6 kinase, Akt/protein kinase B and various protein kinase C isoforms. In VSMCs, AngII also induces the generation of intracellular reactive oxygen species (ROS), which play critical roles in activation and modulation of above signal transduction. Less is known about endothelial cell (EC) AngII signaling than VSMCs, however, recent studies suggest that endothelial AngII signaling negatively regulates the nitric oxide (NO) signaling pathway and thereby induces endothelial dysfunction. Moreover, in both VSMCs and ECs, AngII signaling cross-talk with insulin signaling might be involved in insulin resistance, an important risk factor in the development of cardiovascular diseases. In fact, clinical and pharmacological studies showed that AngII infusion induces insulin resistance and AngII converting enzyme inhibitors and AT(1) receptor blockers improve insulin sensitivity. In this review, we focus on the recent findings that suggest the existence of novel signaling mechanisms whereby AngII mediates processes, such as activation of receptor or non-receptor tyrosine kinases and ROS, as well as cross-talk between insulin and NO signal transduction in VSMCs and ECs.


Subject(s)
Angiotensin II/physiology , Endothelium, Vascular/physiology , Muscle, Smooth, Vascular/physiology , Protein Kinases/physiology , Receptors, Vascular Endothelial Growth Factor/physiology , Signal Transduction/physiology , Angiotensin II/pharmacology , Cardiovascular Diseases/etiology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Muscle, Smooth, Vascular/drug effects , Protein Kinases/metabolism , Receptors, Vascular Endothelial Growth Factor/drug effects , Signal Transduction/drug effects
13.
Article in English | MEDLINE | ID: mdl-16250862

ABSTRACT

Accumulating evidence strongly implicates the critical roles of intracellular signaling of angiotensin II (AngII) in mediating cardiovascular diseases such as hypertension, atherosclerosis, and restenosis after vascular injury. The importance of AngII signals has also been reported in endothelial dysfunction and insulin resistance, two strong predictors of cardiovascular disease. Through its G protein-coupled AngII type-1 receptor (AT1), AngII activates various intracellular protein kinases, such as receptor or non-receptor tyrosine kinases and serine/threonine kinases. Activation of these kinases requires both G protein-dependent and independent pathways, reactive oxygen species and a metalloprotease, and each kinase could be involved specifically in mediating pathophysiological function of the AT1 receptor target organs. In fact, some of the kinases are indispensable for AngII-induced hypertrophy and migration. The role of these AT1-activated kinases in mediating vascular remodeling, vascular contractility, endothelial dysfunction, and insulin resistance will be discussed in this review. In addition, the AT1 receptor undergoes rapid phosphorylation, desensitization, and internalization upon AngII stimulation. Recent studies with site-directed mutagenesis of the AT1 receptor not only elucidated a G protein interaction and desensitization of the receptor, but also demonstrated a structural requirement of the receptor for downstream signal transduction. Thus, AT1 mutants have provided an excellent means to examine the mechanism of signal transduction and their significance in mediating AngII function. Taken together, in this review, we will focus our discussion on the recent findings of the signal transduction research elucidating novel signaling mechanisms of the AT1 receptor that are relevant to the vascular pathophysiology of AngII.


Subject(s)
Angiotensin II/metabolism , Muscle, Smooth, Vascular/physiopathology , Protein Kinases/metabolism , Receptors, Angiotensin/metabolism , Signal Transduction , Humans , Muscle, Smooth, Vascular/metabolism , Receptors, Angiotensin/chemistry , Structure-Activity Relationship
14.
Antioxid Redox Signal ; 7(9-10): 1315-26, 2005.
Article in English | MEDLINE | ID: mdl-16115037

ABSTRACT

Reactive oxygen species (ROS) are proposed to induce cardiovascular diseases, such as atherosclerosis, hypertension, restenosis, and fibrosis, through several mechanisms. One such mechanism involves ROS acting as intracellular second messengers, which lead to induction of unique signal transductions. Angiotensin II (AngII), a potent cardiovascular pathogen, stimulates ROS production through the G protein-coupled AngII type 1 receptor expressed in its target organs, such as vascular tissues, heart, and kidney. Recent accumulating evidence indicates that through ROS production, AngII activates downstream ROS-sensitive kinases that are critical in mediating cardiovascular remodeling. Each of these ROS-sensitive kinases could potentially mediate its own specific function. In this review, we will focus our discussion on the current findings that suggest novel mechanisms of how AngII mediates activation of these redox-sensitive kinases in target organs, as well as the pathological significance of their activation.


Subject(s)
Angiotensin II/metabolism , Gene Expression Regulation, Enzymologic , Oxidation-Reduction , Animals , Fibroblasts/metabolism , Humans , Models, Biological , Reactive Oxygen Species , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Tissue Distribution
15.
Antioxid Redox Signal ; 7(7-8): 1053-61, 2005.
Article in English | MEDLINE | ID: mdl-15998260

ABSTRACT

Although there is an abundance of evidence suggesting that insulin resistance plays a significant role in the vasculature, the precise mechanistic role involved still remains unclear. In this review, we discuss the current background of insulin resistance in the context of insulin signaling and action in the vasculature. Also, studies suggest that insulin resistance, diabetes, and cardiovascular disease all share a common involvement with oxidative stress. Recently, we reported that lysophosphatidylcholine, a major bioactive product of oxidized low-density lipoprotein, and angiotensin II, a vasoactive hormone and a potent inducer of reactive oxygen species (ROS), negatively regulate insulin signaling in vascular smooth muscle cells (VSMCs). In endothelial cells, insulin stimulates the release of nitric oxide, which results in VSMC relaxation and inhibition of atherosclerosis. Other data suggest that angiotensin II inhibits the vasodilator effects of insulin through insulin receptor substrate-1 phosphorylation at Ser312 and Ser616. Moreover, ROS impair insulin-induced vasorelaxation by neutralizing nitric oxide to form peroxynitrite. Thus, evidence is growing to enable us to better understand mechanistically the relationship between insulin/insulin resistance and ROS in the vasculature, and the impact they have on cardiovascular disease.


Subject(s)
Endothelial Cells/metabolism , Insulin/metabolism , Muscle, Smooth, Vascular/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Animals , Humans , Muscle, Smooth, Vascular/blood supply
16.
Arterioscler Thromb Vasc Biol ; 25(9): 1831-6, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15994438

ABSTRACT

BACKGROUND: Rho and its effector Rho-kinase/ROCK mediate cytoskeletal reorganization as well as smooth muscle contraction. Recent studies indicate that Rho and ROCK are critically involved in vascular remodeling. Here, we tested the hypothesis that Rho/ROCK are critically involved in angiotensin II (Ang II)-induced migration of vascular smooth muscle cells (VSMCs) by mediating a specific signal cross-talk. METHODS AND RESULTS: Immunoblotting demonstrated that Ang II stimulated phosphorylation of a ROCK substrate, regulatory myosin phosphatase targeting subunit (MYPT)-1. Phosphorylation of MYPT-1 as well as migration of VSMCs induced by Ang II was inhibited by dominant-negative Rho (dnRho) or ROCK inhibitor, Y27632. Ang II-induced c-Jun NH2-terminal kinase (JNK) activation, but extracellular signal-regulated kinase (ERK) activation was not mediated through Rho/ROCK. Thus, infection of adenovirus encoding dnJNK inhibited VSMC migration by Ang II. We have further demonstrated that the Rho/ROCK activation by Ang II requires protein kinase C-delta (PKCdelta) and proline-rich tyrosine kinase 2 (PYK2) activation, but not epidermal growth factor receptor transactivation. Also, VSMCs express PDZ-Rho guanine nucleotide exchange factor (GEF) and Ang II stimulated PYK2 association with tyrosine phosphorylated PDZ-RhoGEF. CONCLUSIONS: PKCdelta/PYK2-dependent Rho/ROCK activation through PDZ-RhoGEF mediates Ang II-induced VSMC migration via JNK activation in VSMCs, providing a novel mechanistic role of the Rho/ROCK cascade that is involved in vascular remodeling.


Subject(s)
Cell Movement/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Protein Serine-Threonine Kinases/metabolism , rho GTP-Binding Proteins/metabolism , Adenoviridae/genetics , Angiotensin II/pharmacology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cell Movement/drug effects , Cells, Cultured , Focal Adhesion Kinase 2/genetics , Focal Adhesion Kinase 2/metabolism , Gene Expression , Guanine Nucleotide Exchange Factors/metabolism , Intracellular Signaling Peptides and Proteins , Muscle, Smooth, Vascular/drug effects , Protein Kinase C-delta/metabolism , Receptor Cross-Talk/physiology , Rho Guanine Nucleotide Exchange Factors , Vasoconstrictor Agents/pharmacology , rho GTP-Binding Proteins/genetics , rho-Associated Kinases
17.
J Biol Chem ; 280(28): 26592-9, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15905175

ABSTRACT

A G protein-coupled receptor agonist, angiotensin II (AngII), induces epidermal growth factor (EGF) receptor (EGFR) transactivation possibly through metalloprotease-dependent, heparin-binding EGF (HB-EGF) shedding. Here, we have investigated signal transduction of this process by using COS7 cells expressing an AngII receptor, AT1. In these cells AngII-induced EGFR transactivation was completely inhibited by pretreatment with a selective HB-EGF inhibitor, or with a metalloprotease inhibitor. We also developed a COS7 cell line permanently expressing a HB-EGF construct tagged with alkaline phosphatase, which enabled us to measure HB-EGF shedding quantitatively. In the COS7 cell line AngII stimulated release of HB-EGF. This effect was mimicked by treatment either with a phospholipase C activator, a Ca2+ ionophore, a metalloprotease activator, or H2O2. Conversely, pretreatment with an intracellular Ca2+ antagonist or an antioxidant blocked AngII-induced HB-EGF shedding. Moreover, infection of an adenovirus encoding an inhibitor of G(q) markedly reduced EGFR transactivation and HB-EGF shedding through AT1. In this regard, AngII-stimulated HB-EGF shedding was abolished in an AT1 mutant that lacks G(q) protein coupling. However, in cells expressing AT1 mutants that retain G(q) protein coupling, AngII is still able to induce HB-EGF shedding. Finally, the AngII-induced EGFR transactivation was attenuated in COS7 cells overexpressing a catalytically inactive mutant of ADAM17. From these data we conclude that AngII stimulates a metalloprotease ADAM17-dependent HB-EGF shedding through AT1/G(q)/phospholipase C-mediated elevation of intracellular Ca2+ and reactive oxygen species production, representing a key mechanism indispensable for EGFR transactivation.


Subject(s)
Epidermal Growth Factor/metabolism , GTP-Binding Proteins/chemistry , Heparin/chemistry , Metalloproteases/metabolism , Receptor, Angiotensin, Type 2/chemistry , ADAM Proteins , ADAM17 Protein , Adenoviridae/genetics , Alkaline Phosphatase/metabolism , Animals , CHO Cells , COS Cells , Calcium/metabolism , Cricetinae , Epidermal Growth Factor/chemistry , Epidermal Growth Factor/physiology , GTP-Binding Proteins/metabolism , Heparin-binding EGF-like Growth Factor , Hydrogen Peroxide/pharmacology , Intercellular Signaling Peptides and Proteins , Ionophores/pharmacology , Kinetics , Lac Operon , Ligands , Metalloendopeptidases/metabolism , Mutation , Protein Binding , Rats , Reactive Oxygen Species , Retroviridae/genetics , Signal Transduction , Time Factors , Transcriptional Activation , Transfection , Type C Phospholipases/metabolism
18.
Am J Physiol Cell Physiol ; 287(3): C807-13, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15163624

ABSTRACT

Epidermal growth factor (EGF) family ligands have been implicated in cardiovascular diseases because of their enhanced expression in vascular lesions and their promoting effects on growth and migration of vascular smooth muscle cells (VSMCs). Betacellulin (BTC), a novel EGF family ligand, has been shown to be expressed in atherosclerotic lesions and to be a potent growth factor of VSMCs. However, the molecular mechanisms downstream of BTC involved in mediating vascular remodeling remain largely unknown. Therefore, the aim of this study was to examine the effects of BTC on signal transduction, growth, and migration in VSMCs. We found that BTC stimulated phosphorylation of EGF receptor (EGFR) at Tyr1068, which was completely blocked by an EGFR kinase inhibitor, AG-1478. BTC also phosphorylated ErbB2 at Tyr877, Tyr1112, and Tyr1248 and induced association of ErbB2 with EGFR, suggesting their heterodimerization in VSMCs. In postreceptor signal transduction, BTC stimulated phosphorylation of extracellular signal-regulated kinase (ERK)1/2, Akt, and p38 mitogen-activated protein kinase (MAPK). Moreover, BTC stimulated proliferation and migration of VSMCs. ERK and Akt inhibitors suppressed migration markedly and proliferation partially, whereas the p38 inhibitor suppressed migration partially but not proliferation. In addition, we found the presence of endogenous BTC in conditioned medium of VSMCs and an increase of BTC on angiotensin II stimulation. In summary, BTC promotes growth and migration of VSMCs through activation of EGFR, ErbB2, and downstream serine/threonine kinases. Together with the expression and processing of endogenous BTC in VSMCs, our results suggest a critical involvement of BTC in vascular remodeling.


Subject(s)
Cell Movement/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/growth & development , Myocytes, Smooth Muscle/cytology , Signal Transduction/drug effects , Betacellulin , Blotting, Western , Cell Division/drug effects , Cell Movement/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Phosphorylation/drug effects , Precipitin Tests , Receptor, ErbB-2/drug effects , Receptor, ErbB-2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
19.
Antioxid Redox Signal ; 5(6): 771-80, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14588150

ABSTRACT

Enhanced production of reactive oxygen species (ROS) such as H(2)O(2) and a failure in ROS removal by scavenging systems are hallmarks of several cardiovascular diseases such as atherosclerosis and hypertension. ROS act as second messengers that play a prominent role in intracellular signaling and cellular function. In vascular smooth muscle cells (VSMCs), a vascular pathogen, angiotensin II, appears to initiate growth-promoting signal transduction through ROS-sensitive tyrosine kinases. However, the precise mechanisms by which tyrosine kinases are activated by ROS remain unclear. In this review, the current knowledge that suggests how certain tyrosine kinases are activated by ROS, along with their functional significance in VSMCs, will be discussed. Recent findings suggest that transactivation of the epidermal growth factor receptor by ROS requires metalloprotease-dependent heparin-binding epidermal growth factor-like growth factor production, whereas other ROS-sensitive tyrosine kinases such as PYK2, JAK2, and platelet-derived growth factor receptor require activation of protein kinase C-delta. Each of these ROS-sensitive kinases could mediate specific signaling critical for pathophysiological responses. Detailed analysis of the mechanism of cross-talk and the downstream function of these various tyrosine kinases will yield new therapeutic interventions for cardiovascular disease.


Subject(s)
Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Protein-Tyrosine Kinases/metabolism , Reactive Oxygen Species , Animals , Cardiovascular Diseases/pathology , Cells, Cultured , Enzyme Activation , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Humans , Models, Biological , Risk Factors , Signal Transduction , Transcriptional Activation
20.
Exp Biol Med (Maywood) ; 228(7): 836-42, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12876303

ABSTRACT

Both insulin resistance and reactive oxygen species (ROS) have been reported to play essential pathophysiological roles in cardiovascular diseases, such as hypertension and atherosclerosis. However, the mechanistic link between ROS, such as H2O2 and insulin resistance in the vasculature, remains undetermined. Akt, a Ser/Thr kinase, mediates various biological responses induced by insulin. In this study, we examined the effects of H2O2 on Akt activation in the insulin-signaling pathway in vascular smooth muscle cells (VSMCs). In VSMCs, insulin stimulates Akt phosphorylation at Ser473. Pretreatment with H2O2 concentration- and time-dependently inhibited insulin-induced Akt phosphorylation with significant inhibition observed at 50 microM for 10 min. A ROS inducer, diamide, also inhibited insulin-induced Akt phosphorylation. In addition, H2O2 inhibited insulin receptor binding partially and inhibited insulin receptor autophosphorylation almost completely. However, pretreatment with a protein kinase C inhibitor, GF109203X (2 microM), for 30 min did not block the inhibitory effects of H2O2 on insulin-induced Akt phosphorylation, suggesting that protein kinase C is not involved in the inhibition by H2O2. We conclude that ROS inhibit a critical insulin signal transduction component required for Akt activation in VSMCs, suggesting potential cellular mechanisms of insulin resistance, which would require verification in vivo.


Subject(s)
Hydrogen Peroxide/pharmacology , Insulin/pharmacology , Muscle, Smooth, Vascular/metabolism , Protein Serine-Threonine Kinases , Animals , Diamide/pharmacology , Drug Interactions , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Insulin/metabolism , Maleimides/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , Receptor, Insulin/metabolism , Serine/metabolism , Signal Transduction/drug effects , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...