Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Plant Mol Biol ; 58(1): 27-40, 2005 May.
Article in English | MEDLINE | ID: mdl-16028114

ABSTRACT

To identify the gibberellin (GA) signaling components involved in rice leaf sheath elongation process, protein phosphorylation changed by GA3 was analyzed. The protein kinase activities in rice leaf sheath were assessed in an in-gel kinase assay using SDS-polyacrylamide gel containing histone III-S as a substrate. The activity of a putative 54-kDa calcium dependent protein kinase (CDPK) in cytosolic fraction in rice leaf sheath increased significantly by GA3. Further, phosphorylation status of the proteins changed by GA3 in rice leaf sheath were detected by in vitro protein phosphorylation followed by two-dimensional polyacrylamide gel electrophoresis and the phosphoproteins were identified by mass spectrometry. Sixty phosphoproteins was detected after in vitro protein phosphorylation and the phosphorylation of 7 proteins was enhanced by GA3 treatment. The addition of GA3 treated cytosolic fraction of leaf sheath further increased the phosphorylation of 4 proteins, glyoxalase-I, cytoplasmic malate dehydrogenase, glyceraldehydes-3-phosphate dehydrogenase and another unknown protein. The protein kinase inhibitor, staurosporine inhibited the phosphorylation of these proteins in vitro. Other hormones, particularly, indole acetic acid, 6-benzylaminopurine and abscisic acid did not change the phosphorylation status of these proteins. The identified proteins did not show any change by GA3 treatment at transcription level. The abundance of glyoxalase-I and cytoplasmic malate dehydrogenase remained unchanged by GA3 treatment as detected on 2D-gel by silver staining, unlike for glyceraldehydes-3-phosphate dehydrogenase. Results suggest that the phosphoproteins, glyoxalase-I and cytoplasmic malate dehydrogenase in rice leaf sheath could be important signaling components of GA3, downstream to 54-kDa CDPK.


Subject(s)
Gibberellins/pharmacology , Oryza/drug effects , Phosphoproteins/metabolism , Plant Leaves/drug effects , Blotting, Northern , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cytosol/enzymology , Dose-Response Relationship, Drug , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Histones/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Lactoylglutathione Lyase/metabolism , Malate Dehydrogenase/metabolism , Mass Spectrometry , Molecular Weight , Oryza/genetics , Oryza/metabolism , Phosphoproteins/genetics , Phosphorylation/drug effects , Plant Growth Regulators/pharmacology , Plant Leaves/genetics , Plant Leaves/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Plant Structures/drug effects , Plant Structures/genetics , Plant Structures/metabolism , Protein Kinases/chemistry , Protein Kinases/genetics , Protein Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Staurosporine/pharmacology , Substrate Specificity , Time Factors
2.
Circulation ; 112(1): 84-92, 2005 Jul 05.
Article in English | MEDLINE | ID: mdl-15983244

ABSTRACT

BACKGROUND: In the heart, the expressions of several types of prostanoid receptors have been reported. However, their roles in cardiac hypertrophy in vivo remain unknown. We intended to clarify the roles of these receptors in pressure overload-induced cardiac hypertrophy using mice lacking each of their receptors. METHODS AND RESULTS: We used a model of pressure overload-induced cardiac hypertrophy produced by banding of the transverse aorta in female mice. In wild-type mice subjected to the banding, cardiac hypertrophy developed during the observation period of 8 weeks. In mice lacking the prostaglandin (PG) I2 receptor (IP(-/-)), however, cardiac hypertrophy and cardiomyocyte hypertrophy were significantly greater than in wild-type mice at 2 and 4 weeks but not at 8 weeks, whereas there was no such augmentation in mice lacking the prostanoid receptors other than IP. In addition, cardiac fibrosis observed in wild-type hearts was augmented in IP(-/-) hearts, which persisted for up to 8 weeks. In IP(-/-) hearts, the expression level of mRNA for atrial natriuretic peptide, a representative marker of cardiac hypertrophy, was significantly higher than in wild-type hearts. In vitro, cicaprost, an IP agonist, reduced platelet-derived growth factor-induced proliferation of wild-type noncardiomyocytes, although it could not inhibit cardiotrophin-1-induced hypertrophy of cardiomyocytes. Accordingly, cicaprost increased cAMP concentration efficiently in noncardiomyocytes. CONCLUSIONS: IP plays a suppressive role in the development of pressure overload-induced cardiac hypertrophy via the inhibition of both cardiomyocyte hypertrophy and cardiac fibrosis. Both effects have been suggested as originating from the action on noncardiomyocytes rather than cardiomyocytes.


Subject(s)
Cardiomegaly/etiology , Hypertension/complications , Receptors, Epoprostenol/physiology , Animals , Biomarkers/analysis , Cardiomegaly/pathology , Cell Enlargement , Cyclic AMP/blood , Disease Models, Animal , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Female , Fibrosis , Mice , Mice, Knockout , Myocytes, Cardiac/pathology , RNA, Messenger/analysis , Receptors, Epoprostenol/deficiency , Receptors, Epoprostenol/genetics
3.
Nat Med ; 11(5): 562-6, 2005 May.
Article in English | MEDLINE | ID: mdl-15834430

ABSTRACT

Systemic inflammation induces various adaptive responses including tachycardia. Although inflammation-associated tachycardia has been thought to result from increased sympathetic discharge caused by inflammatory signals of the immune system, definitive proof has been lacking. Prostanoids, including prostaglandin (PG) D(2), PGE(2), PGF(2alpha), PGI(2) and thromboxane (TX) A(2), exert their actions through specific receptors: DP, EP (EP(1), EP(2), EP(3), EP(4)), FP, IP and TP, respectively. Here we have examined the roles of prostanoids in inflammatory tachycardia using mice that lack each of these receptors individually. The TXA(2) analog I-BOP and PGF(2alpha) each increased the beating rate of the isolated atrium of wild-type mice in vitro through interaction with TP and FP receptors, respectively. The cytokine-induced increase in beating rate was markedly inhibited in atria from mice lacking either TP or FP receptors. The tachycardia induced in wild-type mice by injection of lipopolysaccharide (LPS) was greatly attenuated in TP-deficient or FP-deficient mice and was completely absent in mice lacking both TP and FP. The beta-blocker propranolol did not block the LPS-induced increase in heart rate in wild-type animals. Our results show that inflammatory tachycardia is caused by a direct action on the heart of TXA(2) and PGF(2alpha) formed under systemic inflammatory conditions.


Subject(s)
Dinoprost/pharmacology , Inflammation , Tachycardia/metabolism , Thromboxane A2/pharmacology , Animals , Blood Pressure , Dinoprost/metabolism , Electrocardiography , Heart Atria/drug effects , Heart Rate/drug effects , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Propranolol/pharmacology , Receptors, Prostaglandin/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Tachycardia/chemically induced
4.
Circulation ; 109(20): 2462-8, 2004 May 25.
Article in English | MEDLINE | ID: mdl-15123528

ABSTRACT

BACKGROUND: In the heart with acute myocardial infarction, production of prostaglandin (PG) E2 increases significantly. In addition, several subtypes of PGE2 receptors (EPs) have been reported to be expressed in the heart. The role of PGE2 in cardiac ischemia-reperfusion (I/R) injury, however, remains unknown. We intended to clarify the role of PGE2 via EP4, an EP subtype, in I/R injury using mice lacking EP4 (EP4-/- mice). METHODS AND RESULTS: In murine cardiac ventricle, competitive reverse transcription-polymerase chain reaction revealed the highest expression level of EP4 mRNA among EP mRNAs. EP4-/- mice had larger infarct size than wild-type mice in a model of I/R; the left anterior descending coronary artery was occluded for 1 hour, followed by 24 hours of reperfusion. In addition, isolated EP4-/- hearts perfused according to the Langendorff technique had greater functional and biochemical derangements in response to I/R than wild-type hearts. In vitro, AE1-329, an EP4 agonist, raised cAMP concentration remarkably in noncardiomyocytes, whereas the action was weak in cardiomyocytes. When 4819-CD, another EP4 agonist, was administered 1 hour before coronary occlusion, it reduced infarct size significantly in wild-type mice. Notably, a similar cardioprotective effect was observed even when it was administered 50 minutes after coronary occlusion. CONCLUSIONS: Both endogenous PGE2 and an exogenous EP4 agonist protect the heart from I/R injury via EP4. The potent cardioprotective effects of 4819-CD suggest that the compound would be useful for treatment of acute myocardial infarction.


Subject(s)
Dinoprostone/physiology , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/prevention & control , Receptors, Prostaglandin E/agonists , Animals , Cardiotonic Agents/therapeutic use , Cyclic AMP/metabolism , Heart Ventricles/metabolism , Male , Methyl Ethers/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , RNA, Messenger/metabolism , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP4 Subtype
5.
Circulation ; 108(19): 2381-6, 2003 Nov 11.
Article in English | MEDLINE | ID: mdl-14557367

ABSTRACT

BACKGROUND: Circulatory failure in sepsis arises from vascular hyporesponsiveness, in which nitric oxide (NO) derived from inducible NO synthase (iNOS) plays a major role. Details of the cross talk between thromboxane (TX) A2 and the iNOS-NO system, however, remain unknown. We intended to clarify the role of TXA2, via the cross talk, in vascular hyporesponsiveness. METHODS AND RESULTS: We examined cytokine-induced iNOS expression and NO production in cultured vascular smooth muscle cells (VSMCs) and cytokine-induced hyporesponsiveness of the aorta from mice lacking the TXA2 receptor (TP-/- mice). The cytokine-induced iNOS expression and NO production observed in wild-type VSMCs were significantly augmented in TP-/- VSMCs, indicating an inhibitory effect of endogenous TXA2 on iNOS expression. Furthermore, in indomethacin-treated wild-type VSMCs, U-46619, a TP agonist, inhibited cytokine-induced iNOS expression and NO production in a concentration-dependent manner, effects absent from TP-/- VSMCs. In an ex vivo system, the cytokine-induced hyporesponsiveness of aortas to phenylephrine was significantly augmented in TP-/- aorta but was almost completely canceled by aminoguanidine, an iNOS inhibitor. Accordingly, cytokine-induced NO production was significantly higher in TP-/- aorta than in wild-type aorta. Moreover, U-46619 significantly suppressed lipopolysaccharide-induced NO production in vivo only in wild-type mice. CONCLUSIONS: These results suggest that TXA2 has a protective role against the development of vascular hyporesponsiveness via its inhibitory action on the iNOS-NO system under pathological conditions such as sepsis.


Subject(s)
Nitric Oxide Synthase/biosynthesis , Thromboxane A2/physiology , Vasoconstrictor Agents/pharmacology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Aorta/drug effects , Cells, Cultured/drug effects , Cells, Cultured/enzymology , Cytokines/pharmacology , Endotoxemia/physiopathology , Enzyme Induction/drug effects , Humans , Indomethacin/pharmacology , Male , Mice , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Nitric Oxide/biosynthesis , Nitric Oxide/physiology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Phenylephrine/pharmacology , Receptors, Thromboxane A2, Prostaglandin H2/deficiency , Receptors, Thromboxane A2, Prostaglandin H2/drug effects , Receptors, Thromboxane A2, Prostaglandin H2/genetics , Thromboxane A2/agonists , Thromboxane A2/pharmacology
6.
Br J Pharmacol ; 136(4): 530-9, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12055131

ABSTRACT

Effects of the prostanoids on the growth of cultured aortic vascular smooth muscle cells (VSMCs) were examined using mice lacking prostanoid receptors. Proliferation of VSMCs was assessed by measuring [(3)H]-thymidine incorporation and the cell number, and their hypertrophy by [(14)C]-leucine incorporation and protein content. In VSMCs from wild-type mice, expressions of mRNAs for the EP(4) and TP were most abundant, followed by those for the IP, EP(3) and FP, when examined by competitive reverse transcriptase-PCR. Those for the EP(1), EP(2) and DP, however, could not be detected. AE1-329, an EP(4) agonist, and cicaprost, an IP agonist, inhibited platelet derived growth factor (PDGF)-induced proliferation of VSMCs from wild-type mice; these inhibitory effects disappeared completely in VSMCs from EP(4)(-/-) and IP(-/-) mice, respectively. In accordance with these effects, AE1-329 and cicaprost stimulated cAMP production in VSMCs from wild-type mice, which were absent in VSMCs from EP(4)(-/-) and IP(-/-) mice, respectively. Effects of PGE(2) on cell proliferation and adenylate cyclase were almost similar with those of AE1-329 in VSMCs from wild-type mice, which disappeared in VSMCs from EP(4)(-/-) mice. PGD(2) inhibited PDGF-induced proliferation of VSMCs from both wild-type and DP(-/-) mice to a similar extent. This action of PGD(2) was also observed in VSMCs from EP4(-/-) and IP(-/-) mice. In VSMCs from wild-type mice, I-BOP, a TP agonist, showed potentiation of PDGF-induced hypertrophy. I-BOP failed to show this action in VSMCs from TP(-/-) mice. The specific agonists for the EP(1), EP(2) or EP(3), and PGF(2)alpha showed little effect on the growth of VSMCs. These results show that PGE(2), PGI(2) and TXA(2) modulate PDGF-induced proliferation or hypertrophy of VSMCs via the EP(4), IP and TP, respectively, and that the inhibitory effect of PGD(2) on PDGF-induced proliferation is not mediated by the DP, EP(4) or IP.


Subject(s)
Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Prostaglandins/pharmacology , Receptors, Prostaglandin/agonists , Animals , Aorta/cytology , Aorta/drug effects , Aorta/pathology , Apoptosis , Cell Count , Cell Division/drug effects , Cells, Cultured , Hypertrophy , Leucine/metabolism , Mice , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Platelet-Derived Growth Factor/pharmacology , Polymerase Chain Reaction , Prostaglandin D2/pharmacology , Prostaglandin D2/physiology , RNA, Messenger/metabolism , Receptors, Prostaglandin/deficiency , Receptors, Prostaglandin/genetics , Thymidine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...