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










Publication year range
1.
J Clin Invest ; 106(7): 907-15, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11018079

ABSTRACT

Chromogranin A (CgA) is the major soluble protein in the core of catecholamine-storage vesicles and is also distributed widely in secretory vesicles throughout the neuroendocrine system. CgA contains the sequences for peptides that modulate catecholamine release, but the proteases responsible for the release of these bioactive peptides from CgA have not been established. We show here that the major fibrinolytic enzyme, plasmin, can cleave CgA to form a series of large fragments as well as small trichloroacetic acid-soluble peptides. Peptides generated by plasmin-mediated cleavage of CgA significantly inhibited nicotinic cholinergic stimulation of catecholamine release from PC12 cells and primary bovine adrenal chromaffin cells. We also show that the zymogen, plasminogen, as well as tissue plasminogen activator bind saturably and with high capacity to catecholaminergic (PC12) cells. Occupancy of cell surface binding sites promoted the cleavage of CgA by plasmin. Positive and negative modulation of the local cellular fibrinolytic system resulted in substantial alterations in catecholamine release. These results suggest that catecholaminergic cells express binding sites that localize fibrinolytic molecules on their surfaces to promote plasminogen activation and proteolytic processing of CgA in the environment into which CgA is secreted to generate peptides which may regulate neuroendocrine secretion. Interactions between CgA and plasmin(ogen) define a previously unrecognized autocrine/paracrine system that may have a dramatic impact upon catecholamine secretion.


Subject(s)
Catecholamines/metabolism , Chromogranins/metabolism , Fibrinolysin/metabolism , Protein Processing, Post-Translational , Adrenal Glands/cytology , Adrenal Glands/metabolism , Animals , Cattle , Cell Communication , Chromaffin Cells/metabolism , Chromogranin A , Fibrinolysis , Lysine/analogs & derivatives , Neurosecretory Systems/metabolism , PC12 Cells , Plasminogen/metabolism , Protein Binding/drug effects , Rats , Receptors, Nicotinic/metabolism , Secretory Vesicles/metabolism , Tissue Plasminogen Activator/metabolism
2.
Invest Ophthalmol Vis Sci ; 41(8): 2309-17, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10892878

ABSTRACT

PURPOSE: To determine the antiangiogenic effects of peroxisome proliferator-activated receptor (PPAR)-gamma agonists on ocular cells involved in the pathogenesis of choroidal neovascularization (CNV) in vitro and on experimental laser photocoagulation-induced CNV in vivo. METHODS: PPAR-gamma expression in human retinal pigment epithelial (RPE) cells and bovine choroidal endothelial cells (CECs) was determined using an RNase protection assay and Western blot analysis. Two PPAR-gamma ligands, troglitazone (TRO) and rosiglitazone (RSG; 0.1-20 microM), were used to assess effects on RPE and CEC proliferation and migration and CEC tube formation in response to vascular endothelial growth factor (VEGF). The effects of intravitreal injection of TRO on laser photocoagulation-induced CNV lesions in rat eyes (15 experimental, 15 control, nine burns per eye) and cynomolgus monkey eyes (two experimental, two control, seven paramacular burns per eye) was assessed by fluorescein angiography and histologic evaluation. RESULTS. PPAR-gamma1 was expressed in both RPE and CEC. PPAR-gamma ligands significantly inhibited VEGF-induced migration and proliferation in both cell types and tube formation of CEC in a dose-response manner. CNV in rats was markedly inhibited by intravitreous injection of TRO (P < 0.001). Lesions showed significantly less fluorescein leakage and were histologically thinner in the TRO-treated animals. Similar findings were present in the TRO-treated lesions in two monkey eyes. The drug showed no apparent adverse effects in the adjacent retina or in control eyes. CONCLUSIONS: The inhibition of VEGF-induced choroidal angiogenesis in vitro, and CNV in vivo by PPAR-gamma ligands suggests the potential application of these agents in the large group of patients with age-related macular degeneration complicated by CNV.


Subject(s)
Choroidal Neovascularization/prevention & control , Chromans/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Thiazoles/pharmacology , Thiazolidinediones , Transcription Factors/metabolism , Animals , Blotting, Western , Cell Division/drug effects , Cell Movement/drug effects , Choroid/blood supply , Choroid/drug effects , Choroid/pathology , Choroidal Neovascularization/etiology , Choroidal Neovascularization/pathology , Chromans/administration & dosage , Dose-Response Relationship, Drug , Endothelial Growth Factors/toxicity , Endothelium, Vascular/metabolism , Fluorescein Angiography , Humans , Injections , Laser Coagulation , Ligands , Lymphokines/toxicity , Macaca fascicularis , Male , Pigment Epithelium of Eye/metabolism , Rats , Rats, Inbred BN , Receptors, Cytoplasmic and Nuclear/agonists , Rosiglitazone , Thiazoles/administration & dosage , Transcription Factors/agonists , Troglitazone , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
3.
J Cardiovasc Pharmacol ; 35(5): 749-57, 2000 May.
Article in English | MEDLINE | ID: mdl-10813377

ABSTRACT

Troglitazone (TRO) is an oral insulin-sensitizer that has direct effects on the vasculature to inhibit cell growth and migration. In vascular smooth muscle cells (VSMCs), insulin transduces a mitogenic signal that is dependent on the ERK1/2 MAP kinases. We examined the effects of TRO on this pathway and found that it inhibits mitogenic signaling. In quiescent VSMCs, insulin (1 microM) induced a 3.2-fold increase in DNA synthesis. TRO (1-20 microM) inhibited insulin-stimulated DNA synthesis by 72.8% at the maximal concentration. TRO at I and 10 microM had no significant effect on insulin-stimulated ERK1/2 activity. At 20 microM, however, TRO modestly enhanced insulin-stimulated ERK1/2 activity by 1.5-fold. ERKs transduce a mitogenic signal by phosphorylating transcription factors such as Elk-1. which regulate critical growth-response genes. We used GAL-Elk-1 expression plasmids to detect ERK-dependent activation of Elk-1. TRO at 1-20 microM potently inhibited insulin-stimulated, ERK1/2-dependent Elk-1 transcription factor activity. Neither early steps in insulin signaling nor the phosphatidylinositol 3-kinase (PI3K) branch of this pathway were affected by TRO, because it had no effect on IRS-1 phosphorylation, PI3K/IRS-1 association, or Akt phosphorylation. Because TRO is a known ligand for the nuclear transcription factor peroxisome proliferator-activated receptor gamma (PPARgamma), we tested two other ligands for this receptor, rosiglitazone (RSG) and 15-deoxy-delta12,14 prostaglandin J2 (15d-PGJ2). Both also inhibited insulin-induced DNA synthesis. In summary, these data show that TRO inhibits mitogenic signaling by insulin at a point distal of ERK1/2 activation, potentially by a PPARgamma-mediated inhibition of ERK-dependent phosphorylation and activation of nuclear transcription factors that regulate cell growth.


Subject(s)
Chromans/pharmacology , DNA-Binding Proteins , Hypoglycemic Agents/pharmacology , Insulin Antagonists/pharmacology , Muscle, Smooth, Vascular/drug effects , Signal Transduction/drug effects , Thiazoles/pharmacology , Thiazolidinediones , Animals , Cells, Cultured , DNA/biosynthesis , DNA/drug effects , Enzyme Activation/drug effects , Fibroblast Growth Factor 2/metabolism , Insulin/metabolism , Insulin Receptor Substrate Proteins , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , Receptor, Insulin/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Troglitazone , ets-Domain Protein Elk-1
4.
Circulation ; 101(11): 1311-8, 2000 Mar 21.
Article in English | MEDLINE | ID: mdl-10725292

ABSTRACT

BACKGROUND: Peroxisome proliferator-activated receptor-gamma (PPARgamma) is activated by fatty acids, eicosanoids, and insulin-sensitizing thiazolidinediones (TZDs). The TZD troglitazone (TRO) inhibits vascular smooth muscle cell (VSMC) proliferation and migration in vitro and in postinjury intimal hyperplasia. METHODS AND RESULTS: Rat and human VSMCs express mRNA and nuclear receptors for PPARgamma1. Three PPARgamma ligands, the TZDs TRO and rosiglitazone and the prostanoid 15-deoxy-Delta(12,14)-prostaglandin J2 (15d-PGJ2), all inhibited VSMC proliferation and migration. PPARgamma is upregulated in rat neointima at 7 days and 14 days after balloon injury and is also present in early human atheroma and precursor lesions. CONCLUSIONS: Pharmacological activation of PPARgamma expressed in VSMCs inhibits their proliferation and migration, potentially limiting restenosis and atherosclerosis. These receptors are upregulated during vascular injury.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , 3T3 Cells/physiology , Animals , Aorta/injuries , Aorta/metabolism , Catheterization , Cell Division/physiology , Cell Movement/physiology , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , DNA/biosynthesis , Fibroblast Growth Factor 2/pharmacology , Humans , Ligands , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Platelet-Derived Growth Factor/pharmacology , RNA, Messenger/metabolism , Rats , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Subcellular Fractions/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tunica Intima/metabolism
5.
Arterioscler Thromb Vasc Biol ; 19(9): 2094-104, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10479650

ABSTRACT

An early event in acute and chronic inflammation and associated diseases such as atherosclerosis and rheumatoid arthritis is the induced expression of specific adhesion molecules on the surface of endothelial cells (ECs), which subsequently bind leukocytes. Peroxisome proliferator-activated receptors (PPARs), members of the nuclear receptor superfamily of transcription factors, are activated by fatty acid metabolites, peroxisome proliferators, and thiazolidinediones and are now recognized as important mediators in the inflammatory response. Whether PPAR activators influence the inflammatory responses of ECs is unknown. We show that the PPAR activators 15-deoxy-Delta(12,14)-prostaglandin J(2) (15d-PGJ(2)), Wyeth 14643, ciglitazone, and troglitazone, but not BRL 49653, partially inhibit the induced expression of vascular cell adhesion molecule-1 (VCAM-1), as measured by ELISA, and monocyte binding to human aortic endothelial cells (HAECs) activated by phorbol 12-myristate 13-acetate (PMA) or lipopolysaccharide. The "natural" PPAR activator 15d-PGJ(2) had the greatest potency and was the only tested molecule capable of partially inhibiting the induced expression of E-selectin and neutrophil-like HL60 cell binding to PMA-activated HAECs. Intracellular adhesion molecule-1 induction by PMA was unaffected by any of the molecules tested. Both PPAR-alpha and PPAR-gamma mRNAs were detected in HAECs by using reverse transcription-polymerase chain reaction and a ribonuclease protection assay; however, we have yet to determine which, if any, of the PPARs are mediating this process. These results suggest that certain PPAR activators may help limit chronic inflammation mediated by VCAM-1 and monocytes without affecting acute inflammation mediated by E-selectin and neutrophil binding.


Subject(s)
Cell Communication/physiology , Endothelium, Vascular/physiology , Leukocytes/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Cell Adhesion/physiology , Cells, Cultured , E-Selectin/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Monocytes/physiology , Neutrophils/physiology , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/pharmacology , Vascular Cell Adhesion Molecule-1/metabolism
6.
FEBS Lett ; 452(3): 277-82, 1999 Jun 11.
Article in English | MEDLINE | ID: mdl-10386606

ABSTRACT

The thiazolidinedione troglitazone inhibits angiotensin II-induced extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase activity in vascular smooth muscle cells. Activation of extracellular signal-regulated kinase 1/2 by angiotensin II is a multistep process involving both its phosphorylation by mitogen-activated protein kinase extracellular signal-regulated kinase kinase in the cytoplasm and a subsequent translocation to the nucleus. The cytoplasmic activation of extracellular signal-regulated kinase 1/2 in vascular smooth muscle cells proceeds through the protein kinase Czeta --> mitogen-activated protein kinase extracellular signal-regulated kinase kinase --> extracellular signal-regulated kinase pathway. Troglitazone did not affect the angiotensin II-induced activation of protein kinase Czeta or its downstream signaling kinases extracellular signal-regulated kinase 1/2 in the cytosol. In contrast, angiotensin II-induced activation of protein kinase Czeta and extracellular signal-regulated kinase 1/2 in the nucleus were both inhibited by troglitazone. Nuclear translocation of extracellular signal-regulated kinase 1/2 induced by angiotensin II was completely blocked by troglitazone. Protein kinase Czeta, however, did not translocate upon angiotensin II stimulation. Troglitazone, therefore, inhibits both angiotensin II-induced nuclear translocation of extracellular signal-regulated kinase 1/2 and the nuclear activity of its upstream signaling kinase protein kinase Czeta. Since extracellular signal-regulated kinase 1/2 nuclear translocation may be a critical signaling step for multiple growth factors that stimulate vascular smooth muscle cells proliferation and migration, troglitazone may provide a new therapeutical approach for the prevention and treatment of atherosclerosis and restenosis.


Subject(s)
Angiotensin II/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Nucleus/metabolism , Chromans/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases , Muscle, Smooth, Vascular/metabolism , Thiazoles/pharmacology , Thiazolidinediones , Vasodilator Agents/pharmacology , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Cell Nucleus/drug effects , Cells, Cultured , Cytoplasm/metabolism , Kinetics , Mitogen-Activated Protein Kinase 3 , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Troglitazone
7.
J Cardiovasc Pharmacol ; 33(5): 798-806, 1999 May.
Article in English | MEDLINE | ID: mdl-10226869

ABSTRACT

The purpose of this study was to determine the effect of the peroxisome proliferator-activated receptor gamma-(PPAR gamma) ligands troglitazone (TRO), rosiglitazone (RSG), and 15-deoxy-delta prostaglandin J2 (15d-PGJ2) on vascular smooth muscle cell (VSMC) migration directed by multiple chemoattractants. Involvement of mitogen-activated protein kinase (MAPK) in migration also was examined, because TRO was previously shown to inhibit nuclear events stimulated by this pathway during mitogenic signaling in VSMCs. Migration of rat aortic VSMCs was induced 5.4-fold by PDGF, 4.6-fold by thrombin, and 2.3-fold by insulin-like growth factor I (IGF-I; all values of p < 0.05). The PPAR gamma ligands 15d-PGJ2, RSG, or TRO all inhibited VSMC migration with the following order of potency: 15d-PGJ2 > RSG > TRO. Inhibition of MAPK signaling with PD98059 completely blocked PDGF-, thrombin-, and IGF-I-induced migration. All chemoattractants induced MAPK activation. PPAR gamma ligands did not inhibit MAPK activation, suggesting a nuclear effect of these ligands downstream of MAPK. The importance of nuclear events was confirmed because actinomycin D also blocked migration. We conclude that PPAR gamma ligands are potent inhibitors of VSMC migration pathways, dependent on MAPK and nuclear events. PPAR gamma ligands act downstream of the cytoplasmic activation of MAPK and appear to exert their effects in the nucleus. Because VSMC migration plays an important role in the formation of atherosclerotic lesions and restenosis, PPAR gamma ligands like TRO and RSG, which ameliorate insulin resistance in humans, also may protect the vasculature from diabetes-enhanced injury.


Subject(s)
Aorta, Thoracic/physiology , Cell Movement , Chemotactic Factors/physiology , Muscle, Smooth, Vascular/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Thiazolidinediones , Transcription Factors/physiology , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Movement/drug effects , Cells, Cultured , Chemotactic Factors/pharmacology , Chromans/pharmacology , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Enzyme Activation , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor I/physiology , Ligands , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Platelet-Derived Growth Factor/pharmacology , Platelet-Derived Growth Factor/physiology , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/pharmacology , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/drug effects , Rosiglitazone , Thiazoles/pharmacology , Thrombin/pharmacology , Thrombin/physiology , Transcription Factors/drug effects , Troglitazone
8.
Hypertension ; 33(1 Pt 2): 183-9, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9931102

ABSTRACT

-Migration of vascular smooth muscle cells (VSMC) is a key event in neointimal formation and atherosclerosis that may be linked to the accumulation of inflammatory cells and release of chemotactic cytokines. Tumor necrosis factor-alpha (TNF-alpha) induces chemotaxis of inflammatory cells and fibroblasts, but little is known about chemotactic signaling by TNF-alpha in VSMC. The aim of this study was to investigate the role of TNF-alpha in VSMC migration and to elucidate the chemotactic signaling pathways mediating this action. TNF-alpha (50 to 400 U/mL) induced migration of cultured rat aortic VSMC in a dose-dependent manner. Because activation of the extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase (MAPK) is known to be required in platelet-derived growth factor-directed and angiotensin II-directed migration of these cells, we used the MAPK-inhibitor PD98059 to determine if chemotactic signaling by TNF-alpha involves the MAPK pathway as well. We found that TNF-alpha-directed migration was substantially inhibited by PD98059. TNF-alpha (100 U/mL) transiently activated MAPK with a maximal induction 10 minutes after stimulation that returned to baseline levels by 2 hours after treatment. Only a single peak of increased MAPK activity was seen. PD98059 also blocked TNF-alpha-stimulated MAPK activation in a concentration-dependent manner, which is consistent with its inhibition of TNF-alpha-directed migration. To identify which TNF-alpha receptor is involved in TNF-alpha-induced MAPK activation, antibodies against the p55 TNF-alpha receptor-1 (TNF-R1) and the p75 TNF-alpha receptor-2 (TNF-R2) were used. VSMC express both receptors, but TNF-alpha-induced MAPK activation was inhibited only by the TNF-R1 antibody. The TNF-R2 antibody had no effect. Thiazolidinediones are known to inhibit TNF-alpha signaling in adipose tissue and attenuate platelet-derived growth factor-directed and angiotensin II-directed migration in VSMC. We therefore investigated the effects of the thiazolidinediones troglitazone (TRO) and rosiglitazone (RSG) on TNF-alpha-induced migration. Both TRO and RSG inhibited migration, but neither attenuated TNF-alpha-induced MAPK activation, indicating that their antimigration activity was exerted downstream of MAPK. These experiments provide the first evidence that early activation of MAPK is a crucial event in TNF-alpha-mediated signal transduction leading to VSMC migration. Moreover, inhibition of TNF-alpha-directed migration by the insulin sensitizers TRO and RSG underscores their potential as vasculoprotective agents.


Subject(s)
Aorta, Thoracic/physiology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Chemotaxis/physiology , Mitogen-Activated Protein Kinases , Muscle, Smooth, Vascular/physiology , Tumor Necrosis Factor-alpha/pharmacology , Analysis of Variance , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Cells, Cultured , Chemotaxis/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Kinetics , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Tumor Necrosis Factor-alpha/physiology
9.
Arterioscler Thromb Vasc Biol ; 19(1): 73-82, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9888869

ABSTRACT

Angiotensin II (Ang II) promotes vascular smooth muscle cell (VSMC) growth and migration, but the signaling pathways mediating these VSMC behaviors critical to restenosis and atherosclerosis are not completely known. The purpose of the present investigation was to define the role of mitogen-activated protein kinase (MAPK) in Ang II-induced DNA synthesis, migration, and c-fos induction in VSMCs. PD 98059, a synthetic inhibitor of MAPK kinase, or antisense oligodeoxynucleotides (ODNs) to deplete extracellular signal-regulated kinase (ERK)1 and ERK2 MAPKs, were used to inhibit MAPK signaling. PD 98059 at 30 micromol/L reduced Ang II-induced MAPK activity by 69% (P<0.01). Under these conditions, Ang II-induced DNA synthesis was completely inhibited (P<0.01), and Ang II-directed migration was attenuated by 76% (P<0.05). In contrast, induction of c-fos by Ang II was only partially suppressed (58% inhibition, P<0.01). Antisense ODNs against the initiation site of rat ERK1 and ERK2 MAPK mRNAs reduced corresponding protein levels by 63% (P<0.01) and completely inhibited MAPK activation by either Ang II (1 micromol/L) or 10% serum. Antisense ODNs (0.4 micromol/L) completely inhibited Ang II-induced DNA synthesis (P<0.01), decreased migration by 47% (P<0.01), and reduced c-fos induction by 40% (P<0.01 versus control ODN-transfected VSMCs). The Ang II type 1 (AT1)-receptor blocker irbesartan completely blocked DNA synthesis, migration, MAPK activation, and c-fos induction by Ang II in VSMCs. These results demonstrate that activation of MAPK plays a crucial role in Ang II-directed migration and DNA synthesis through the AT1 receptor. In contrast, Ang II-mediated c-fos induction and migration were only partially inhibited by either antisense ODNs or PD 98059, suggesting that other pathways in addition to the MAPK pathway may be involved in these actions of Ang II. We conclude that MAPK is a critical regulatory factor for Ang II-mediated migration and growth in VSMCs. Ang II-induced DNA synthesis showed a stronger MAPK dependence than did Ang II-directed migration or c-fos induction.


Subject(s)
Angiotensin II/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Movement , DNA/biosynthesis , Mitogen-Activated Protein Kinases , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Movement/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Oligodeoxyribonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-fos/biosynthesis , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/physiology , Signal Transduction/drug effects
10.
FEBS Lett ; 417(3): 283-6, 1997 Nov 17.
Article in English | MEDLINE | ID: mdl-9409734

ABSTRACT

Insulin-stimulated DNA synthesis, MAP kinase (MAPK) activity and c-fos expression in vascular smooth muscle cells (VSMCs) was blocked by the MAPK inhibitor PD 98059. Regulation of c-fos expression by the transcription factor Elk-1 at the serum response element (SRE) is dependent on its phosphorylation by MAPK. PD 98059 also suppressed insulin-induced Elk-1 transcriptional activity through the SRE. These data show that MAPK plays a critical role in both insulin-mediated growth and Elk-1-dependent induction of c-fos in VSMCs.


Subject(s)
Aorta, Thoracic/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , DNA-Binding Proteins , Insulin/pharmacology , Muscle, Smooth, Vascular/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins/metabolism , Transcriptional Activation/physiology , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cells, Cultured , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Genes, fos/drug effects , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , ets-Domain Protein Elk-1
11.
Hypertension ; 29(1 Pt 2): 334-9, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9039124

ABSTRACT

Migration of vascular smooth muscle cells (VSMCs) is a crucial response to vascular injury resulting in neointima formation and atherosclerosis. Platelet-derived growth factor (PDGF-BB) functions as a potent chemoattractant for VSMCs and enhances these pathologies in the vasculature. However, little is known about the intracellular pathways that mediate VSMC migration. In the present study, we investigated the role of mitogen-activated protein kinase (MAPK) activation in this function, since PDGF-BB as well as other growth factors activate this pathway. Using an in-gel kinase assay, we observed that PD 98059 an inhibitor of MEK that activates MAP kinase, inhibited PDGF-BB-induced activation of ERK-1 and ERK-2 in cultured rat aortic smooth muscle cells in a concentration-dependent manner. In contrast, PDGF-mediated activation of intracellular calcium release was not affected by PD 98059. The chemotactic response of both rat aortic smooth muscle cells (RASMCs) and human umbilical vein smooth muscle cells (HUSMCs) toward PDGF-BB (10 ng/mL) was significantly reduced by PD 98059 (10 mumol/L) to 41.7 +/- 7.1% in RASMCs (P < .01) and to 47.2 +/- 5.3% in HUSMCs (P < .01). Similar inhibition was seen at 30 mumol/L, less at 1 mumol/L. To further confirm the specificity of these results implicating the MAPK pathway, an antisense oligodeoxynucleotide (ODN) directed against the initiation translation site of rat ERK-1 and ERK-2 mRNA was used to suppress MAP kinase synthesis and function in rat VSMCs. Liposomal transfection with 0.4 mumol/L antisense ODN reduced ERK-1 and ERK-2 protein by 65% (P < .01) after 48 hours. The chemotactic response to PDGF-BB (10 ng/mL) was reduced by 75% (P < .01) in rat VSMCs transfected with the same antisense ODN concentration. Sense and scrambled control ODNs (0.4 mumol/L) did not affect ERK-1 and ERK-2 protein concentrations or chemotaxis of VSMCs induced by PDGF-BB. These experiments provide the first evidence that activation of MAPK is a critical event in PDGF-mediated signal transduction regulating VSMC migration.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Movement/drug effects , Flavonoids/pharmacology , Muscle, Smooth, Vascular/drug effects , Platelet-Derived Growth Factor/pharmacology , Animals , Becaplermin , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Movement/physiology , Down-Regulation , Enzyme Activation , Humans , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Oligonucleotides, Antisense/pharmacology , Platelet-Derived Growth Factor/antagonists & inhibitors , Proto-Oncogene Proteins c-sis , RNA, Messenger , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
12.
FEBS Lett ; 400(1): 119-21, 1997 Jan 02.
Article in English | MEDLINE | ID: mdl-9000525

ABSTRACT

Angiotensin II (AII) plays a crucial role in controlling the proliferation and migration of vascular smooth muscle cells (VSMCs). The present study was undertaken to determine if troglitazone (Tro) has an effect on the G-protein coupled signaling through AII type I (AT-1) receptors in cultured rat aortic VSMCs. AII-induced MAP kinase activation was inhibited 67.9% by Tro. AII-induced DNA synthesis and migration was completely inhibited by Tro or by the AT-1 receptor blocker irbesartan. The present study demonstrates that troglitazone inhibits AII-induced DNA synthesis, migration and MAP kinase activation in VSMCs which are important molecular events for the development of neointimal hyperplasia and atherosclerosis.


Subject(s)
Angiotensin II/antagonists & inhibitors , Chromans/pharmacology , DNA/biosynthesis , Hypoglycemic Agents/pharmacology , Muscle, Smooth, Vascular/drug effects , Thiazoles/pharmacology , Thiazolidinediones , Vasoconstrictor Agents/antagonists & inhibitors , Angiotensin II/pharmacology , Animals , Aorta, Thoracic , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Movement , Cells, Cultured , DNA/drug effects , Enzyme Activation , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Rats , Troglitazone , Vasoconstrictor Agents/pharmacology
13.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 16(11): 661-3, 1996 Nov.
Article in Chinese | MEDLINE | ID: mdl-9772616

ABSTRACT

Thirty six patients of pulmonary or nasopharyngeal carcinoma were treated with Xilixin granule (XLXG) combined with radiotherapy and compared their efficacy with that of 31 patients treated by Zhenqi Fuzheng granule combined with radiotherapy for control. Results showed that the symptoms of Yin Deficiency syndrome in treated group were obviously improved, the leucocyte decreased by 5.6%, while in control group it reached 25.8%, the 3 year survival rate was significantly higher in treated group (75.0%) than that in control group (51.6%). Animal experiment revealed that XLXG had the effects of tumor inhibition, it could increase white blood cells, platelets and hemoglobin of patients, especially in using large dosage. These results suggested that XLXG have some protective effect against radiotherapeutic damage in patients with malignant tumor.


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Leukopenia/prevention & control , Lung Neoplasms/drug therapy , Nasopharyngeal Neoplasms/drug therapy , Adolescent , Adult , Aged , Animals , Combined Modality Therapy , Female , Humans , Leukocyte Count , Lung Neoplasms/mortality , Lung Neoplasms/radiotherapy , Male , Mice , Middle Aged , Nasopharyngeal Neoplasms/mortality , Nasopharyngeal Neoplasms/radiotherapy , Sarcoma 180/drug therapy , Survival Rate
14.
J Clin Invest ; 98(8): 1897-905, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8878442

ABSTRACT

Vascular smooth muscle cell (VSMC) proliferation and migration are responses to arterial injury that are highly important to the processes of restenosis and atherosclerosis. In the arterial balloon injury model in the rat, platelet-derived growth factor (PDGF) and basic fibroblast growth factor (bFGF) are induced in the vessel wall and regulate these VSMC activities. Novel insulin sensitizing agents, thiazolidinediones, have been demonstrated to inhibit insulin and epidermal growth factor-induced growth of VSMCs. We hypothesized that these agents might also inhibit the effect of PDGF and bFGF on cultured VSMCs and intimal hyperplasia in vivo. Troglitazone (1 microM), a member of the thiazolidinedione class, produced a near complete inhibition of both bFGF-induced DNA synthesis as measured by bromodeoxyuridine incorporation (6.5+/-3.9 vs. 17.6+/-4.3% cells labeled, P < 0.05) and c-fos induction. This effect was associated with an inhibition (by 73+/-4%, P < 0.01) by troglitazone of the transactivation of the serum response element, which regulates c-fos expression. Inhibition of c-fos induction by troglitazone appeared to occur via a blockade of the MAP kinase pathway at a point downstream of MAP kinase activation by MAP kinase kinase. At this dose, troglitazone also inhibited PDGF-BB-directed migration of VSMC (by 70+/-6%, P < 0.01). These in vitro effects were operative in vivo. Quantitative image analysis revealed that troglitazone-treated rats had 62% (P < 0.001) less neointima/media area ratio 14 d after balloon injury of the aorta compared with injured rats that received no troglitazone. These results suggest troglitazone is a potent inhibitor of VSMC proliferation and migration and, thus, may be a useful agent to prevent restenosis and possibly atherosclerosis.


Subject(s)
Chromans/pharmacology , Hypoglycemic Agents/pharmacology , Muscle, Smooth, Vascular/drug effects , Thiazoles/pharmacology , Thiazolidinediones , Animals , Arteriosclerosis/prevention & control , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , DNA/biosynthesis , Fibroblast Growth Factor 2/antagonists & inhibitors , Genes, fos/drug effects , Hyperplasia , Male , Muscle, Smooth, Vascular/pathology , Platelet-Derived Growth Factor/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Troglitazone
15.
Kidney Int ; 50(3): 745-53, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8872947

ABSTRACT

Angiotensin II (Ang II) and insulin are implicated in the mesangial cell hypertrophy and excessive accumulation of mesangial matrix seen in glomerulosclerosis. Therefore, the effects of Ang II with and without insulin on mRNA levels of several important extracellular matrix genes and transforming growth factor beta-1 (TGF-beta 1) were examined. Ang II alone (1 microM) added to quiescent, murine mesangial cells in serum-free, insulin-free media slightly but not significantly increased TGF-beta 1, fibronectin, collagen I, collagen IV and laminin message levels. The slight elevations in message expression were reversed by losartan, suggesting that these modest effects are mediated by the AT-1 receptor. Ang II alone also had no significant effects on TGF-beta 1 and extracellular matrix message levels in quiescent rat mesangial cells. In contrast, significant increases in mRNA for collagen 1 (6-fold), collagen IV (4-fold), fibronectin 1 (4-fold) and TGF-beta 1 (2-fold) were seen with insulin alone (10(-6)M) in rat mesangial cells, and a dose-response effect could be demonstrated for insulin (10(-9) to 10(-6)M). Ang II plus insulin further significantly increased collagen I (9-fold), collagen IV (9-fold), fibronectin 1 (5-fold) and TGF-beta 1 (3-fold) message expression. These effects were partially reversed in the presence of losartan. The Northern analyses were supported by measurements of active and total TGF-beta 1 activity (pg/ml/ 5 x 10(6) cells): 1145 +/- 76 and 1960 +/- 199, serum free control; 1121 +/- 92 and 1932 +/- 214, Ang II (10(-6)M); 4589 +/- 103 (P < 0.001 vs. control) and 11071 +/- 1952 (P < 0.01 vs. control), insulin (10(-6)M); and 6881 +/- 183 (P < 0.001 vs. control) and 16626 +/- 1435 (P < 0.01 vs. control), insulin plus Ang II. These results suggest that insulin, itself, significantly increases TGF-beta 1 and extracellular matrix gene expression in rat mesangial cells. Ang II alone has modest effects, while Ang II and insulin have additive effects. To explain the mechanism of these additive effects, we investigated the action of Ang II on insulin signaling and the effect of insulin on Ang II AT1 receptor mRNA expression. Ang II did not enhance insulin-induced insulin receptor substrate-1 (IRS-1) phosporylation or phosphatidylinositol3 (PI-3) kinase activity, but did enhance insulin-induced mitogen activated protein (MAP) kinase activity. Insulin increased message levels of AT1 receptor by twofold. These results suggest that enhancement of MAP kinase activity and AT1 receptor regulation by insulin may contribute to the additive effects of insulin and Ang II in mesangial cells.


Subject(s)
Angiotensin II/pharmacology , Extracellular Matrix Proteins/genetics , Glomerular Mesangium/drug effects , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Transforming Growth Factor beta/genetics , Vasoconstrictor Agents/pharmacology , Animals , Blotting, Northern , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Collagen/genetics , Drug Interactions/physiology , Extracellular Matrix Proteins/metabolism , Fibronectins/genetics , Gene Expression/drug effects , Glomerular Mesangium/cytology , Glomerular Mesangium/enzymology , Mice , Procollagen/genetics , Proto-Oncogene Proteins/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
16.
J Clin Invest ; 92(2): 1042-54, 1993 Aug.
Article in English | MEDLINE | ID: mdl-8394383

ABSTRACT

Secretory proteins are targeted into either constitutive (secreted upon synthesis) or regulated (stored in vesicles and released in response to a secretagogue) pathways. To investigate mechanisms of protein targeting into catecholamine storage vesicles (CSV), we stably expressed human chromogranin A (CgA), the major soluble protein in human CSV, in the rat pheochromocytoma PC-12 cell line. Chromaffin cell secretagogues (0.1 mM nicotinic cholinergic agonist, 55 mM K+, or 2 mM Ba++) caused cosecretion of human CgA and catecholamines from human CgA-expressing cells. Sucrose gradients colocalized human CgA and catecholamines to subcellular particles of the same buoyant density. Chimeric proteins, in which human CgA (either full-length [457 amino acids] or truncated [amino-terminal 226 amino acids]) was fused in-frame to the ordinarily nonsecreted protein chloramphenicol acetyltransferase (CAT), were expressed transiently in PC-12 cells. Both constructs directed CAT activity into regulated secretory vesicles, as judged by secretagogue-stimulated release. These data demonstrate that human CgA expressed in PC-12 cells is targeted to regulated secretory vesicles. In addition, human CgA can divert an ordinarily non-secreted protein into the regulated secretory pathway, consistent with the operation of a dominant targeting signal for the regulated pathway within the peptide sequence of CgA.


Subject(s)
Chromogranins/metabolism , Cytoplasmic Granules/metabolism , Norepinephrine/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Avian Sarcoma Viruses/genetics , Base Sequence , Carbachol/pharmacology , Chloramphenicol O-Acetyltransferase/biosynthesis , Chloramphenicol O-Acetyltransferase/metabolism , Chromogranin A , Chromogranins/biosynthesis , Chromogranins/genetics , Cytoplasmic Granules/ultrastructure , Genetic Vectors , Humans , Microscopy, Electron , Molecular Sequence Data , Oligodeoxyribonucleotides , PC12 Cells , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Transfection
17.
Neurochem Int ; 22(4): 361-7, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8457772

ABSTRACT

Evidence suggests that proenkephalin and members of the chromogranin/secretogranin family of proteins are prohormone precursors, giving rise to a variety of peptides with biologic activity. However, the specific proteases responsible for cleaving these proteins in vivo have not been fully established. Several candidate proteases have been described, some of which have been shown to cleave these proteins in vitro. Proteolytic processing of the chromogranins may be particularly complex, occurring in specific tissue-dependent patterns. To account for this level of complexity several protease systems may be operative, either alone or in concert, both within the neurosecretory granule and in the extracellular space. Specific proteases which are available within neurosecretory cells or in the local extracellular environment, and which may cleave these prohormones include PC1 and PC2 (recently described members of the Kex2/furin family of endoproteases), as well as kallikrein, acetylcholinesterase, and, more recently, the plasminogen/plasmin protease system. The potential role of these specific proteases in the processing of proenkephalin and the chromogranins is discussed, in particular, in the context of possible processing clues available from recent analysis of cDNA and genomic intron/exon structure.


Subject(s)
Chromaffin Granules/metabolism , Endopeptidases/metabolism , Protein Processing, Post-Translational , Proteins/metabolism , Animals , Chromogranins/genetics , Chromogranins/metabolism , Enkephalins/genetics , Enkephalins/metabolism , Exons , Humans , Protein Precursors/genetics , Protein Precursors/metabolism , Proteins/genetics
18.
Blood ; 78(2): 479-87, 1991 Jul 15.
Article in English | MEDLINE | ID: mdl-1648983

ABSTRACT

The goal of the present study was to assess the relative importance of receptor-bound and secreted plasminogen activator urokinase (u-PA) in generating cell-surface plasmin and fostering destruction of normal tissue by tumor cells. We first showed that active site-inhibited u-PA could displace endogenous u-PA from the surface of the human colon adenocarcinoma cell line HCT 116. We then prepared expression vectors for u-PA and for a mutant molecule in which the codon for the active site serine residue was changed to encode alanine. Expression of non-functional mutant u-PA decreased the level of cell-bound active u-PA by more than 95% via a mechanism that involved competition for receptor sites. Decreased cell-surface u-PA activity was associated with a decrease in cell-bound plasmin activity to undetectable levels, suggesting that receptor-bound u-PA plays an important role in the generation of plasmin on the cell surface. Transfectants that secreted eightfold to 20-fold elevated levels of active wild-type u-PA showed approximately 50% increases in cell-associated u-PA and only twofold to fourfold increases in cell-associated plasmin, suggesting that the role of secreted u-PA in generating cell-surface plasmin activity was relatively minor. In parent cells and both types of transfectants there was a good correlation between the amount of plasmin bound to the tumor cell surface and the extent to which a basement membrane substrate was degraded. These studies show that receptor-bound u-PA provides an efficient mechanism for plasmin generation on the surface of tumor cells, which, in turn, contributes significantly to their degradative potential.


Subject(s)
Fibrinolysin/biosynthesis , Plasminogen Activators/metabolism , Receptors, Cell Surface/physiology , Urokinase-Type Plasminogen Activator/metabolism , Adenocarcinoma , Base Sequence , Basement Membrane/metabolism , Binding Sites , Cell Line , Colonic Neoplasms , Genetic Vectors , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligonucleotide Probes , Plasminogen Activators/genetics , RNA, Messenger/genetics , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Receptors, Urokinase Plasminogen Activator , Transfection , Urokinase-Type Plasminogen Activator/genetics
19.
J Clin Invest ; 87(2): 747-51, 1991 Feb.
Article in English | MEDLINE | ID: mdl-1846882

ABSTRACT

The mammalian proximal tubule is an important mediator of the renal adaptive response to systemic acidosis. In chronic metabolic and respiratory acidosis the bicarbonate reabsorptive (or proton secretory) capacity is increased. This increase is mediated, at least in part, by an increase in Vmax of the luminal Na/H antiporter. To determine whether this adaptation involves increased mRNA expression, Na/H antiporter mRNA levels were measured by Northern analysis in renal cortex of rats with metabolic (6 mmol/kg body wt NH4Cl for 2 or 5 d) and respiratory (10% CO2/air balanced for 2 or 5 d) acidosis and of normal, pair-fed rats. Na/H antiporter mRNA levels were unchanged after 2 d of both metabolic and respiratory acidosis. After 5 d, however, Na/H antiporter mRNA expression was increased 1.76 +/- 0.12-fold in response to metabolic acidosis (P less than 0.005, n = 8), but was not different from normal in response to respiratory acidosis: 1.1 +/- 0.2 (NS, n = 8). Thus, the renal adaptive response to metabolic acidosis involves increased cortical Na/H antiporter mRNA levels. In contrast, the enhanced proximal tubule Na/H antiporter activity and bicarbonate reabsorption in respiratory acidosis seem to involve mechanisms other than increased Na/H antiporter gene expression.


Subject(s)
Acidosis, Respiratory/metabolism , Acidosis/metabolism , Carrier Proteins/genetics , Kidney/metabolism , RNA, Messenger/analysis , Animals , Blotting, Northern , DNA Probes , Gene Expression , Male , Nucleic Acid Hybridization , Rats , Rats, Inbred Strains , Sodium-Hydrogen Exchangers
20.
Hepatology ; 9(2): 302-6, 1989 Feb.
Article in English | MEDLINE | ID: mdl-2912830

ABSTRACT

Centrilobular hypoxia mediated by enhanced hepatic consumption of oxygen has been hypothesized to be a factor of pathogenetic importance in ethanol-induced liver injury. In the present study, this hypothesis was tested in a rat model which developed alcoholic centrilobular liver necrosis. Male Wistar rats were infused with high fat diet plus ethanol or isocaloric glucose for 7 weeks, a duration which resulted in induction of balloon cell degeneration, focal necrosis, and inflammation in the centrilobular region of the liver of the ethanol-fed animals. Hepatic blood flow, oxygen consumption and oxygen delivery were determined by the radiolabeled microsphere method and measurement of oxygen content in arterial, portal venous, and hepatic venous blood. Hepatic oxygen consumption was markedly increased by 159% in the ethanol-fed animals compared to that in the controls when results were expressed as relative to body weight. Even after these results were standardized per gram of liver weight, hepatic oxygen consumption was still significantly elevated in the ethanol-fed group, but the magnitude of the elevation was reduced to 70%, due to marked hepatomegaly observed in these animals. There was a concomitant 59% increase in hepatic oxygen delivery in the ethanol-fed rats when expressed per kilogram of body weight, and this effect was attributable entirely to increased portal blood flow. However, the increment of this increase in oxygen delivery was much too small to compensate for the 159% increase in oxygen consumption. In addition, this increase in hepatic oxygen delivery was no longer observable when the results were reexpressed based on the liver weight.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Liver Diseases, Alcoholic/metabolism , Liver/metabolism , Oxygen Consumption , Animals , Body Weight , Hepatic Veins , Liver/pathology , Liver Circulation , Liver Diseases, Alcoholic/pathology , Male , Necrosis , Organ Size , Oxygen/blood , Rats , Rats, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL
...