Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 284
Filter
1.
Cell Mol Life Sci ; 61(11): 1317-30, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15170510

ABSTRACT

Ribosome-catalyzed peptide bond formation is a crucial function of all organisms. The ribosome is a ribonucleoprotein particle, with both RNA and protein components necessary for the various steps leading to protein biosynthesis. Evolutionary theory predicts an early environment devoid of complex biomolecules, and prebiotic peptide synthesis would have started in a simple way. A fundamental question regarding peptide synthesis is how the current ribosome-catalyzed reaction evolved from a primitive system. Here we look at both prebiotic and modern mechanisms of peptide bond formation and discuss recent experiments that aim to connect these activities. In particular, RNA can facilitate peptide bond formation by providing a template for activated amino acids to react and can catalyze a variety of functions that would have been necessary in a pre-protein world. Therefore, RNA may have facilitated the emergence of the current protein world from an RNA or even prebiotic world.


Subject(s)
Biological Evolution , Peptides , Protein Biosynthesis , Catalysis , RNA, Ribosomal , RNA, Transfer , Ribosomes/chemistry , Ribosomes/physiology
4.
J Biol Chem ; 276(51): 48269-75, 2001 Dec 21.
Article in English | MEDLINE | ID: mdl-11585822

ABSTRACT

Angiotensin II (Ang II) induces transactivation of the epidermal growth factor (EGF) receptor (EGF-R), which serves as a scaffold for various signaling molecules in vascular smooth muscle cells (VSMCs). Cholesterol and sphingomyelin-enriched lipid rafts are plasma membrane microdomains that concentrate various signaling molecules. Caveolae are specialized lipid rafts that are organized by the cholesterol-binding protein, caveolin, and have been shown to be associated with EGF-Rs. Angiotensin II stimulation promotes a rapid movement of AT(1) receptors to caveolae; however, their functional role in angiotensin II signaling has not been elucidated. Here we show that cholesterol depletion by beta-cyclodextrin disrupts caveolae structure and concomitantly inhibits tyrosine phosphorylation of the EGF-R and subsequent activation of protein kinase B (PKB)/Akt induced by angiotensin II. Similar inhibitory effects were obtained with other cholesterol-binding agents, filipin and nystatin. In contrast, EGF-R autophosphorylation and activation of Akt/PKB in response to EGF are not affected by cholesterol depletion. The early Ang II-induced upstream signaling events responsible for transactivation of the EGF-R, such as the intracellular Ca(2+) increase and c-Src activation, also remain intact. The EGF-R initially binds caveolin, but these two proteins rapidly dissociate following angiotensin II stimulation during the time when EGF-R transactivation is observed. The activated EGF-R is localized in focal adhesions together with tyrosine-phosphorylated caveolin. These findings suggest that 1) a scaffolding role of caveolin is essential for EGF-R transactivation by angiotensin II and 2) cholesterol-rich microdomains as well as focal adhesions are important signal-organizing compartments required for the spatial and temporal organization of angiotensin II signaling in VSMCs.


Subject(s)
Angiotensin II/physiology , Cholesterol/metabolism , ErbB Receptors/genetics , Muscle, Smooth, Vascular/physiology , Protein Serine-Threonine Kinases , Transcriptional Activation , beta-Cyclodextrins , Animals , Caveolin 1 , Caveolins/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Cyclodextrins/pharmacology , Enzyme Activation , Epidermal Growth Factor/physiology , ErbB Receptors/chemistry , Fluorescent Antibody Technique , Male , Microscopy, Electron , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/ultrastructure , Phosphorylation , Protein Binding , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , Signal Transduction , Tyrosine/metabolism
5.
Article in English | MEDLINE | ID: mdl-11550797

ABSTRACT

Aminoacyl-tRNA synthetases are modular proteins, with domains that have distinct roles in the aminoacylation reaction. The catalytic core is responsible for aminoacyl adenylate formation and transfer of the amino acid to the 3' end of the bound transfer RNA (tRNA). Appended and inserted domains contact portions of the tRNA outside the acceptor site and contribute to the efficiency and specificity of aminoacylation. Some aminoacyl-tRNA synthetases also have distinct editing activities that are localized to unique domains. Efficient aminoacylation and editing require communication between RNA-binding and catalytic domains, and can be considered as a signal transduction system. Here, evidence for domain-domain communication in aminoacyl-tRNA synthetases is summarized, together with insights from structural analysis.


Subject(s)
Amino Acyl-tRNA Synthetases/chemistry , Amino Acyl-tRNA Synthetases/metabolism , Amino Acyl-tRNA Synthetases/genetics , Base Sequence , Escherichia coli/enzymology , Escherichia coli/genetics , Molecular Sequence Data , Nucleic Acid Conformation , Protein Structure, Tertiary , RNA Editing , RNA, Transfer, Amino Acyl/genetics , RNA, Transfer, Amino Acyl/metabolism
7.
Arterioscler Thromb Vasc Biol ; 21(4): 489-95, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11304462

ABSTRACT

Angiotensin II (Ang II) is a vasoactive hormone with critical roles in vascular smooth muscle cell growth, an important feature of hypertension and atherosclerosis. Many of these effects are dependent on the production of reactive oxygen species (ROS). Ang II induces phosphorylation of the epidermal growth factor (EGF) receptor (EGF-R), which serves as a scaffold for various signaling molecules. Here, we provide novel evidence that ROS are critical mediators of EGF-R transactivation by Ang II. Pretreatment of vascular smooth muscle cells with the antioxidants diphenylene iodonium, Tiron, N-acetylcysteine, and ebselen significantly inhibited ( approximately 80% to 90%) tyrosine phosphorylation of the EGF-R by Ang II but not by EGF. Of the 5 autophosphorylation sites on the EGF-R, Ang II mainly phosphorylated Tyr1068 and Tyr1173 in a redox-sensitive manner. The Src family kinase inhibitor PP1, overexpression of kinase-inactive c-Src, or chelation of intracellular Ca(2+) attenuated EGF-R transactivation. Although antioxidants had no effects on the Ca(2+) mobilization or phosphorylation of Ca(2+)-dependent tyrosine kinase Pyk2, they inhibited c-Src activation by Ang II, suggesting that c-Src is 1 signaling molecule that links ROS and EGF-R phosphorylation. Furthermore, Ang II-induced tyrosine phosphorylation of the autophosphorylation site and the SH2 domain of c-Src was redox sensitive. These findings emphasize the importance of ROS in specific Ang II-stimulated growth-related signaling pathways and suggest that redox-sensitive EGF-R transactivation may be a potential target for antioxidant therapy in vascular disease.


Subject(s)
Angiotensin II/physiology , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Reactive Oxygen Species/metabolism , 1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt/pharmacology , Angiotensin II/pharmacology , Animals , Antioxidants/pharmacology , Azoles/pharmacology , ErbB Receptors/physiology , Isoindoles , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Onium Compounds/pharmacology , Organoselenium Compounds/pharmacology , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/physiology , Receptor, ErbB-2/drug effects , Receptor, ErbB-2/physiology , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , Tyrosine/metabolism
10.
Trends Biochem Sci ; 25(9): 427, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10986456
12.
Arterioscler Thromb Vasc Biol ; 20(2): 385-91, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10669634

ABSTRACT

Monocyte chemoattractant protein-1 (MCP-1) is an important component of the inflammatory response of the vessel wall and has been shown to be regulated by cytokines, such as tumor necrosis factor-alpha (TNF-alpha). However, the precise signaling pathways leading to MCP-1 induction have not been fully elucidated in vascular smooth muscle cells (VSMCs). Cytokine signal transduction involves protein kinases as well as reactive oxygen species (ROS). The relation between these 2 factors is not clear. In this study, we show that TNF-alpha induces a parallel phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) and p38 mitogen-activated protein kinase (p38MAPK) and increases MCP-1 mRNA expression in cultured VSMCs. Inhibition of ERK1/2 but not p38MAPK caused a partial attenuation of MCP-1 induction (43+/-10% inhibition). Incubation of VSMCs with multiple antioxidants (diphenylene iodonium, liposomal superoxide dismutase, catalase, N-acetylcysteine, dimethylthiourea, and pyrrolidine dithiocarbamate) had no effect on TNF-alpha-mediated MCP-1 upregulation. However, simultaneous blockade of the ERK1/2 and ROS pathways by using PD098059 combined with diphenylene iodonium or N-acetylcysteine potently enhanced the ability of MAPK kinase inhibitors to abrogate MCP-1 mRNA expression (100+/-2% inhibition). Thus, parallel ROS-dependent and ERK1/2-dependent pathways converge to regulate TNF-alpha-induced MCP-1 gene expression in VSMCs. These data unmask a complex but organized integration of ROS and protein kinases that mediates cytokine-induced vascular inflammatory gene expression.


Subject(s)
Chemokine CCL2/metabolism , Mitogen-Activated Protein Kinases/physiology , Muscle, Smooth, Vascular/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Cells, Cultured , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Oxidation-Reduction , Rats , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation
13.
Mol Pharmacol ; 57(3): 460-7, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10692485

ABSTRACT

The effects of angiotensin II (Ang II) are mediated primarily by Ang II type 1 receptors, which in turn are coupled to heterotrimeric G proteins. After receptor activation, the G(alpha) and G(betagamma) subunits dissociate, contributing to the signaling cascades involving protein kinase C (PKC) activation. Regulators of G protein signaling (RGS proteins) comprise a class of proteins that have been shown to negatively regulate the G(alpha) subunit. We examined which RGS sequences were expressed in vascular smooth muscle cells and which of these were regulated by Ang II. Reverse transcription-polymerase chain reaction showed that of 16 RGS sequences screened, six RGS transcripts (RGS2, 3, 10, 11, and 12 and GAIP) were present. Northern blot analysis demonstrated that RGS3, 10, and 12 and GAIP were not regulated by Ang II at the mRNA level. In contrast, RGS2 mRNA was rapidly and dose dependently increased (395 +/- 24% peak, 45 min) by Ang II but returned to baseline level by 6 to 8 h. Phorbol-12-myristate-13-acetate, a PKC activator, robustly increased RGS2. This signal was attenuated by the PKC inhibitor GF 109203X (50 +/- 4%) and by phorbol-12, 13-dibutyrate-mediated down-regulation of PKC (48 +/- 13%). Tyrosine kinase inhibition and calcium deprivation did not affect the up-regulation of RGS2 mRNA after Ang II stimulation. Actinomycin D treatment inhibited both Ang II- and phorbol-12-myristate-13-acetate-stimulated RGS2 up-regulation, suggesting activation of transcription by these agonists. The stability of RGS2 mRNA did not appear to be affected by Ang II. Thus, RGS2 is a likely candidate for negative regulation of the G proteins coupled to the Ang II type 1 receptor in vascular smooth muscle cells. Regulation of this protein may be of critical importance in modulating the role of Ang II in vascular disease.


Subject(s)
Angiotensin II/physiology , Gene Expression Regulation , Muscle, Smooth, Vascular/metabolism , RGS Proteins/genetics , Animals , Calcium/metabolism , Cells, Cultured , Male , Muscle, Smooth, Vascular/cytology , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , RGS Proteins/biosynthesis , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Receptors, Angiotensin/drug effects , Receptors, Angiotensin/metabolism , Signal Transduction/drug effects , Up-Regulation
14.
Biochemistry ; 38(49): 16359-65, 1999 Dec 07.
Article in English | MEDLINE | ID: mdl-10587461

ABSTRACT

We report here evidence for mutations that break domain-domain functional communication in a synthetase-tRNA complex. Each synthetase is roughly divided into two major domains that are paralleled by the two arms of the L-shaped tRNA structure. The active-site-containing domain interacts with the acceptor arm of the tRNA. The second domain frequently interacts with the anticodon-containing arm. By an induced-fit mechanism, contacts with the anticodon can activate formation of a robust transition state at a site over 70 A away. This induced-fit-based activation is thought to occur through domain-domain signaling and is seen by the enhancement of aminoacylation of the anticodon-containing full tRNA versus a substrate based on the acceptor arm alone. Here we describe a rationally designed mutant methionyl-tRNA synthetase containing two point substitutions at sites that potentially link an anticodon-binding motif to the catalytic domain. The double mutation had no effect on interactions with either the isolated acceptor arm or the anticodon stem-loop. In contrast to interactions with the separate pieces, the mutant enzyme was severely impaired for binding the native tRNA and lost much of its ability to enhance the rate of charging of the full tRNA over that of a substrate based on the acceptor arm alone. We propose that these residues are part of a network for facilitating domain-domain communication for formation of an active synthetase-tRNA complex by induced fit.


Subject(s)
Methionine-tRNA Ligase/metabolism , Acylation , Alanine/genetics , Amino Acid Substitution/genetics , Anticodon/metabolism , Asparagine/genetics , Base Sequence , Binding Sites/genetics , Enzyme Activation/genetics , Escherichia coli/enzymology , Gene Expression Regulation, Enzymologic , Kinetics , Methionine/metabolism , Methionine-tRNA Ligase/genetics , Methionine-tRNA Ligase/isolation & purification , Methionine-tRNA Ligase/physiology , Molecular Sequence Data , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , RNA, Transfer, Met/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Thermus thermophilus/enzymology
15.
Circulation ; 100(11): 1223-9, 1999 Sep 14.
Article in English | MEDLINE | ID: mdl-10484544

ABSTRACT

BACKGROUND: Cardiovascular ischemic events may occur more frequently in hypertensive patients with activated renin-angiotensin systems. We tested the hypothesis that angiotensin II (Ang II) may contribute to atherosclerosis by increasing expression of vascular inflammatory genes such as vascular cell adhesion molecule-1 (VCAM-1). METHODS AND RESULTS: Rats infused with norepinephrine or Ang II for 6 days developed similar hypertensive responses, but only Ang II-treated rats exhibited significant increases in aortic VCAM-1 protein and mRNA expression. Oral losartan treatment (50 mg. kg(-1). d(-1)) inhibited Ang II-induced hypertension and aortic VCAM-1 mRNA expression. Ang II treatment significantly increased VCAM-1 mRNA expression in cultured rat aortic smooth muscle cells (RASMCs). Ang II also induced nuclear NF-kappaB-like binding activity and transactivated an NF-kappaB-driven VCAM-1 promoter. Losartan and proteasome inhibitors blocked Ang II-induced NF-kappaB activation and VCAM-1 mRNA accumulation. IkappaB-alpha overexpression in RASMCs inhibited Ang II-induced VCAM-1 promoter transactivation. CONCLUSIONS: Ang II may contribute to atherogenesis by activation of VCAM-1 through proteasome dependent, NF-kappaB-like transcriptional mechanisms.


Subject(s)
Angiotensin II/pharmacology , Arteriosclerosis/physiopathology , Renin-Angiotensin System/physiology , Vascular Cell Adhesion Molecule-1/biosynthesis , Animals , Blood Pressure/drug effects , Cells, Cultured , Gene Expression/drug effects , Hypertension/physiopathology , Immunohistochemistry , Male , Norepinephrine/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Angiotensin/physiology , Vascular Cell Adhesion Molecule-1/genetics
16.
J Biol Chem ; 274(32): 22699-704, 1999 Aug 06.
Article in English | MEDLINE | ID: mdl-10428852

ABSTRACT

Angiotensin II, a hypertrophic/anti-apoptotic hormone, utilizes reactive oxygen species (ROS) as growth-related signaling molecules in vascular smooth muscle cells (VSMCs). Recently, the cell survival protein kinase Akt/protein kinase B (PKB) was proposed to be involved in protein synthesis. Here we show that angiotensin II causes rapid phosphorylation of Akt/PKB (6- +/- 0.4-fold increase). Exogenous H(2)O(2) (50-200 microM) also stimulates Akt/PKB phosphorylation (maximal 8- +/- 0.2-fold increase), suggesting that Akt/PKB activation is redox-sensitive. Both angiotensin II and H(2)O(2) stimulation of Akt/PKB are abrogated by the phosphatidylinositol 3-kinase (PI3-K) inhibitors wortmannin and LY294002 (2(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one), suggesting that PI3-K is an upstream mediator of Akt/PKB activation in VSMCs. Furthermore, diphenylene iodonium, an inhibitor of flavin-containing oxidases, or overexpression of catalase to block angiotensin II-induced intracellular H(2)O(2) production significantly inhibits angiotensin II-induced Akt/PKB phosphorylation, indicating a role for ROS in agonist-induced Akt/PKB activation. In VSMCs infected with dominant-negative Akt/PKB, angiotensin II-stimulated [(3)H]leucine incorporation is attenuated. Thus, our studies indicate that Akt/PKB is part of the remarkable spectrum of angiotensin II signaling pathways and provide insight into the highly organized signaling mechanisms coordinated by ROS, which mediate the hypertrophic response to angiotensin II in VSMCs.


Subject(s)
Angiotensin II/pharmacology , Hypertrophy/etiology , Muscle, Smooth, Vascular/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Reactive Oxygen Species/metabolism , Animals , Aorta, Thoracic/cytology , Enzyme Activation , Hydrogen Peroxide/pharmacology , Male , Muscle, Smooth, Vascular/cytology , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Onium Compounds/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , Signal Transduction
17.
Clin Cardiol ; 22(6): 385-90, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10376176

ABSTRACT

This paper extols the value of combining two systems in order to improve learning, teaching, communication, patient care, and clinical research in patients with heart disease. This is accomplished by using Weed's recommendation regarding the creation of a complete Problem List and, within this context, characterizing the cardiac or vascular problem according to the recommendations of the New York Heart Association.


Subject(s)
Cardiovascular Diseases/classification , Medical History Taking , Cardiovascular Diseases/diagnosis , Humans , New York , Societies, Medical
18.
J Mol Biol ; 286(2): 521-40, 1999 Feb 19.
Article in English | MEDLINE | ID: mdl-9973568

ABSTRACT

Nucleotides 518-533 form a loop in ribosomal 30 S subunits that is almost universally conserved. Both biochemical and genetic evidence clearly implicate the 530 loop in ribosomal function, with respect both to the accuracy control mechanism and to tRNA binding. Here, building on earlier work, we identify proteins and nucleotides (or limited sequences) site-specifically photolabeled by radioactive photolabile oligoDNA probes targeted toward the 530 loop of 30 S subunits. The probes we employ are complementary to 16 S rRNA nucleotides 517-527, and have aryl azides attached to nucleotides complementary to nucleotides 518, 522, and 525-527, positioning the photogenerated nitrene a maximum of 19-26 A from the complemented rRNA base. The crosslinks obtained are used as constraints to revise an earlier model of 30 S structure, using the YAMMP molecular modeling package, and to place the 530 loop region within that structure.


Subject(s)
Escherichia coli/ultrastructure , Models, Molecular , Nucleic Acid Conformation , RNA, Bacterial/chemistry , RNA, Ribosomal, 16S/chemistry , Ribosomes/ultrastructure , Cross-Linking Reagents/radiation effects , DNA, Complementary/metabolism , Escherichia coli/chemistry , Photoaffinity Labels/radiation effects , RNA, Bacterial/radiation effects , RNA, Messenger/chemistry , RNA, Messenger/radiation effects , RNA, Ribosomal, 16S/radiation effects , Ribosomal Proteins/chemistry , Ribosomal Proteins/radiation effects , Ribosomes/chemistry , Ribosomes/metabolism
19.
Mol Pharmacol ; 55(1): 142-9, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9882708

ABSTRACT

In cultured vascular smooth muscle cells (VSMCs), activation of phospholipase D (PLD) by angiotensin II (Ang II) represents a major source of sustained generation of second messengers. Understanding the molecular mechanisms controlling activation of this pathway is essential to clarify the complexities of Ang II signaling, but the most proximal mechanisms coupling AT1 receptors to PLD have not been defined. Here we examine the role of heterotrimeric G proteins in AT1 receptor-PLD coupling. In alpha-toxin permeabilized VSMCs, GTPgammaS enhanced Ang II-stimulated PLD activation. In intact cells, Ang II activation of PLD was pertussis toxin-insensitive and was not additive with sodium fluoride, a cell-permeant activator of heterotrimeric G proteins, indicating that AT1 receptor-PLD coupling requires pertussis toxin-insensitive heterotrimeric G proteins. Ang II-stimulated PLD activity was significantly inhibited in VSMCs electroporated with anti-Gbeta antibody (56 +/- 5%) and in cells overexpressing the Gbetagamma-binding region of the carboxyl terminus of beta-adrenergic receptor kinase1 (79 +/- 8%), suggesting a critical role for Gbetagamma in PLD activation by Ang II. This effect may be mediated by pp60(c-src), because in beta-adrenergic receptor kinase1 overexpressing cells, pp60(c-src) activation was inhibited, and in normal cells anti-pp60(c-src) antibody inhibited Ang II-stimulated PLD activity. Galpha12 may also contribute to AT1 receptor-PLD coupling because electroporation of anti-Galpha12 antibody significantly inhibited PLD activity, whereas anti-Galphai and Galphaq/11 antibodies had no effect. Furthermore, electroporation of anti-RhoA antibody also attenuated Ang II-induced PLD activation, suggesting a role for small molecular weight G protein RhoA in this response. Thus, we provide evidence here that Gbetagamma as well as Galpha12 subunits mediate AT1 receptor coupling to tonic PLD activation via pp60(c-src)-dependent mechanisms, and that RhoA is involved in these signaling pathways in rat VSMCs. These results may provide insight into the molecular mechanisms underlying the highly organized, complex, chronic signaling programs associated with vascular smooth muscle growth and remodeling in response to Ang II.


Subject(s)
GTP-Binding Proteins/physiology , Muscle, Smooth, Vascular/enzymology , Phospholipase D/metabolism , Receptors, Angiotensin/physiology , Angiotensin II/pharmacology , Animals , Cells, Cultured , Enzyme Activation , GTP-Binding Proteins/chemistry , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Male , Proto-Oncogene Proteins pp60(c-src)/physiology , Rats , Rats, Sprague-Dawley , rhoA GTP-Binding Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...