Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Am J Physiol Renal Physiol ; 309(11): F980-90, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26447218

ABSTRACT

Diabetic nephropathy (DN) is the main cause of morbidity and mortality in diabetes and is characterized by mesangial matrix deposition and podocytopathy, including podocyte loss. The risk factors and mechanisms involved in the pathogenesis of DN are still not completely defined. In the present study, we aimed to understand the cellular mechanisms through which activation of B2 kinin receptors contribute to the initiation and progression of DN. Stimulation of cultured rat podocytes with bradykinin (BK) resulted in a significant increase in ROS generation, and this was associated with a significant increase in NADPH oxidase (NOX)1 and NOX4 protein and mRNA levels. BK stimulation also resulted in a signicant increase in the phosphorylation of ERK1/2 and Akt, and this effect was inhibited in the presence of NOX1 and Nox4 small interfering (si)RNA. Furthermore, podocytes stimulated with BK resulted in a significant increase in protein and mRNA levels of connective tissue growth factor (CTGF) and, at the same time, a significant decrease in protein and mRNA levels of nephrin. siRNA targeted against NOX1 and NOX4 significantly inhibited the BK-induced increase in CTGF. Nephrin expression was increased in response to BK in the presence of NOX1 and NOX4 siRNA, thus implicating a role for NOXs in modulating the BK response in podocytes. Moreover, nephrin expression in response to BK was also significantly increased in the presence of siRNA targeted against CTGF. These findings provide novel aspects of BK signal transduction pathways in pathogenesis of DN and identify novel targets for interventional strategies.


Subject(s)
Bradykinin/pharmacology , Connective Tissue Growth Factor/metabolism , Membrane Proteins/metabolism , Podocytes/drug effects , Animals , Cells, Cultured , Connective Tissue Growth Factor/genetics , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Membrane Proteins/genetics , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidase 1 , NADPH Oxidase 4 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Phosphorylation , Podocytes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Messenger/metabolism , Rats , Receptor, Bradykinin B2/agonists , Receptor, Bradykinin B2/metabolism , Signal Transduction/drug effects , Time Factors , Transfection , Up-Regulation
2.
J Biol Regul Homeost Agents ; 28(4): 587-603, 2014.
Article in English | MEDLINE | ID: mdl-25620170

ABSTRACT

The contribution of plasma prekallikrein (PK) to vascular remodeling is becoming increasingly recognized. Plasma PK is activated when the zymogen PK is digested to an active enzyme by activated factor XII (FXII). Here, we present our findings that vascular smooth muscle cells (VSMC) activate plasma PK in the absence of FXII. Extracted plasma membrane and cytosolic fractions of VSMCs activate PK, but the rate of PK activation was greater by the membrane fraction. FXII neutralizing antibody did not affect PK activation by extracted proteins of VSMCs. VSMC PKA was inhibited by the serine protease inhibitors such as aprotinin, phenylmethylsulfonyl fluoride, leupeptin and CTI with CI50 of 0.78 µM, 1 mM, 3.13 µM and 40 nM on the cultured cells, respectively. No inhibition of PK activation by cysteine, aspartic acid, and metalloprotease inhibitors was observed. This is the first report of the presence of an intrinsic PKA in VSMC. Considering that VSMCs are normally separated from the circulating blood by endothelial cells, direct PK activation by VSMCs may play a role in disease states like diabetes, hyperlipidemia or hypertension where the endothelial layer is damaged.


Subject(s)
Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Prekallikrein/metabolism , Angiotensin II/pharmacology , Animals , Bradykinin/pharmacology , Cells, Cultured , Humans , Male , Muscle, Smooth, Vascular/cytology , Protease Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley
3.
J Pept Res ; 59(4): 139-48, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11972749

ABSTRACT

Bradykinin (BK) is an endogenous peptide that has been implicated in several pathological conditions, hence antagonists of its activity have therapeutic potential. The decapeptide Hoe 140 is currently one of the best BK antagonists, but interest remains in finding even more potent compounds. A library of Hoe 140 derivatives was synthesized that incorporated non-natural analogs of the cationic, naturally occurring amino acids arginine (Arg) and lysine (Lys). The modified amino acids were designed to form enhanced ionic interactions due to an increase in local hydrophobicity, which promotes desolvation of the cation in water. The potencies of the resulting peptides were determined by competitive binding assays in human A431 cells expressing the BK B2 receptor. Two of the peptides synthesized were equipotent to Hoe 140 (IC(50s) 2.99 and 3.36 nM) and the most potent was demonstrated as a functional antagonist in vitro by blocking BK-mediated phosphorylation of mitogen-activated protein (MAP) kinases. The new derivatives are more hydrophobic than Hoe 140 and thus may exhibit changes in pharmacokinetic properties when evaluated in vivo.


Subject(s)
Bradykinin Receptor Antagonists , Bradykinin/analogs & derivatives , Bradykinin/chemistry , Bradykinin/chemical synthesis , Arginine/analogs & derivatives , Binding, Competitive , Bradykinin/metabolism , Bradykinin/pharmacology , Cell Line , Humans , Kinetics , Lysine/analogs & derivatives , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Receptor, Bradykinin B2 , Receptors, Bradykinin/metabolism
4.
Hypertension ; 38(4): 858-63, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11641299

ABSTRACT

Obese hypertensives have increased nonesterified fatty acids (NEFAs) and alpha-adrenergic vascular reactivity. Raising NEFAs locally with intralipid and heparin augments dorsal hand venoconstrictor responses to phenylephrine, an alpha(1)-adrenoceptor agonist. The enhanced venoconstrictor responses were reversed by indomethacin. The findings suggest that raising NEFAs leads to the generation of cyclooxygenase (COX) product(s) that enhance vascular reactivity. To test this notion, 6-keto-PGF(1alpha) and TxB(2), the stable metabolites of prostaglandin H(2) (PGH(2)); prostacyclin (PGI(2)); and thromboxane (TxA(2)), were measured approximately 1.5 to 2 cm downstream of a dorsal hand vein infusion of intralipid and heparin (n=10) or saline and heparin (n=5) for 2 hours each. During the third hour, intralipid and heparin (experimental) and saline and heparin (control) were continued, and either saline (control) or indomethacin (intervention) were infused. Intralipid and heparin raised local 6-keto PGF(1alpha) concentrations by 350% to 500% (P<0.005), but saline and heparin did not (P=NS). TxB(2) levels did not change significantly with any infusion. Infusion of indomethacin during the third hour of intralipid and heparin lowered plasma 6-keto-PGF(1alpha) (P<0.05), whereas infusion of saline with intralipid and heparin did not (P=NS). Oleic and linoleic acids at 100 micromol/L, increased 6-keto-PGF(1alpha) in vascular smooth muscle cells (VSMCs) through a protein kinase C and extracellular, signal-regulated kinase independent pathway. However, oleic and linoleic acids increased intracellular Ca(2+) in VSMCs. The data indicate that NEFAs induce the production of COX products, perhaps via Ca(2+)-dependent activation of phospholipase A(2). The COX product(s) may contribute to increased vascular alpha-adrenergic reactivity among insulin-resistant individuals when NEFAs are elevated.


Subject(s)
6-Ketoprostaglandin F1 alpha/blood , Fat Emulsions, Intravenous/pharmacology , Hand/blood supply , Veins/drug effects , 6-Ketoprostaglandin F1 alpha/metabolism , Adult , Animals , Calcium/metabolism , Cells, Cultured , Female , Heparin/pharmacology , Humans , Indomethacin/pharmacology , Linoleic Acid/pharmacology , Male , Middle Aged , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Oleic Acid/pharmacology , Oleic Acids , Rats , Stearic Acids/pharmacology , Thromboxane B2/blood , Time Factors
5.
Hypertension ; 38(3 Pt 2): 602-5, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11566939

ABSTRACT

Vascular smooth muscle cell (VSMC) proliferation is a prominent feature of the atherosclerotic process that occurs after endothelial injury. Although a vascular wall kallikrein-kinin system has been described, its contribution to vascular disease remains undefined. Because the B(1)-kinin receptor subtype (B1KR) is induced in VSMCs only in response to injury, we hypothesize that this receptor may be mediating critical events in the progression of vascular disease. In the present study, we provide evidence that des-Arg(9)-bradykinin (dABK) (10(-8) M), acting through B1KR, stimulates the phosphorylation of mitogen-activated protein kinase (MAPK) (p42(mapk) and p44(mapk)). Activation of MAPK by dABK is mediated via a cholera toxin-sensitive pathway and appears to involve protein kinase C, Src kinase, and MAPK kinase. These findings demonstrate that the activation of B1KR in VSMCs leads to the generation of second messengers that converge to activate MAPK and provide a rationale to investigate the mitogenic actions of dABK in vascular injury.


Subject(s)
Bradykinin/analogs & derivatives , Muscle, Smooth, Vascular/metabolism , Receptors, Bradykinin/metabolism , Animals , Bradykinin/pharmacology , Cells, Cultured , Cholera Toxin/pharmacology , Enzyme Activation/drug effects , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Indoles/pharmacology , Interleukin-1/pharmacology , Male , Maleimides/pharmacology , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Phosphorylation/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Bradykinin B1 , Receptors, Bradykinin/drug effects , Receptors, Bradykinin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Virulence Factors, Bordetella/pharmacology
6.
J Appl Physiol (1985) ; 91(3): 1412-20, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11509543

ABSTRACT

A high concentration of circulating low-density lipoproteins (LDL) is a major risk factor for atherosclerosis. Native LDL and LDL modified by glycation and/or oxidation are increased in diabetic individuals. LDL directly stimulate vascular smooth muscle cell (VSMC) proliferation; however, the mechanisms remain undefined. The extracellular signal-regulated kinase (ERK) pathway mediates changes in cell function and growth. Therefore, we examined the cellular effects of native and modified LDL on ERK phosphorylation in VSMC. Addition of native, mildly modified (oxidized, glycated, glycoxidized) and highly modified (highly oxidized, highly glycoxidized) LDL at 25 microg/ml to rat VSMC for 5 min induced a fivefold increase in ERK phosphorylation. To elucidate the signal transduction pathway by which LDL phosphorylate ERK, we examined the roles of the Ca(2+)/calmodulin pathway, protein kinase C (PKC), src kinase, and mitogen-activated protein kinase kinase (MEK). Treatment of VSMC with the intracellular Ca(2+) chelator EGTA-AM (50 micromol/l) significantly increased ERK phosphorylation induced by native and mildly modified LDL, whereas chelation of extracellular Ca(2+) by EGTA (3 mmol/l) significantly reduced LDL-induced ERK phosphorylation. The calmodulin inhibitor N-(6-aminohexyl)-1-naphthalenesulfonamide (40 micromol/l) significantly decreased ERK phosphorylation induced by all types of LDL. Downregulation of PKC with phorbol myristate acetate (5 micromol/l) markedly reduced LDL-induced ERK phosphorylation. Pretreatment of VSMC with a cell-permeable MEK inhibitor (PD-98059, 40 micromol/l) significantly decreased ERK phosphorylation in response to native and modified LDL. These findings indicate that native and mildly and highly modified LDL utilize similar signaling pathways to phosphorylate ERK and implicate a role for Ca(2+)/calmodulin, PKC, and MEK. These results suggest a potential link between modified LDL, vascular function, and the development of atherosclerosis in diabetes.


Subject(s)
Lipoproteins, LDL/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/enzymology , Animals , Aorta/cytology , Arteriosclerosis/metabolism , Calcium/metabolism , Calmodulin/metabolism , Cytoplasm/enzymology , Diabetic Angiopathies/metabolism , Enzyme Activation/drug effects , Humans , Lipoproteins, LDL/metabolism , MAP Kinase Signaling System/physiology , Male , Muscle, Smooth, Vascular/cytology , Phosphorylation , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley
7.
J Biol Chem ; 276(20): 17339-46, 2001 May 18.
Article in English | MEDLINE | ID: mdl-11278760

ABSTRACT

We used a cultured murine cell model of the inner medullary collecting duct (mIMCD-3 cells) to examine the regulation of the ubiquitous sodium-proton exchanger, Na+/H+ exchanger isoform 1 (NHE-1), by a prototypical G protein-coupled receptor, the bradykinin B2 receptor. Bradykinin rapidly activates NHE-1 in a concentration-dependent manner as assessed by proton microphysiometry of quiescent cells and by 2'-7'-bis[2-carboxymethyl]-5(6)-carboxyfluorescein fluorescence measuring the accelerated rate of pH(i) recovery from an imposed acid load. The activation of NHE-1 is blocked by inhibitors of the bradykinin B2 receptor, phospholipase C, Ca2+/calmodulin (CaM), and Janus kinase 2 (Jak2), but not by pertussis toxin or by inhibitors of protein kinase C and phosphatidylinositol 3'-kinase. Immunoprecipitation studies showed that bradykinin stimulates the assembly of a signal transduction complex that includes CaM, Jak2, and NHE-1. CaM appears to be a direct substrate for phosphorylation by Jak2 as measured by an in vitro kinase assay. We propose that Jak2 is a new indirect regulator of NHE-1 activity, which modulates the activity of NHE-1 by increasing the tyrosine phosphorylation of CaM and most likely by increasing the binding of CaM to NHE-1.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins , Receptors, Bradykinin/physiology , Sodium-Hydrogen Exchangers/metabolism , Animals , Bradykinin/pharmacology , Cell Line , Enzyme Inhibitors/pharmacology , Hydrogen-Ion Concentration , Janus Kinase 2 , Kidney Medulla/cytology , Kidney Medulla/physiology , Kinetics , Mice , Mice, Transgenic , Models, Biological , Pertussis Toxin , Receptor, Bradykinin B2 , Receptors, Bradykinin/genetics , Signal Transduction/physiology , Simian virus 40/genetics , Sodium/metabolism , Transcription, Genetic , Type C Phospholipases/metabolism , Virulence Factors, Bordetella/pharmacology
8.
Am J Physiol Heart Circ Physiol ; 280(4): H1537-46, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11247764

ABSTRACT

The development of vascular disease is accelerated in hyperglycemic states. Vascular injury plays a pivotal role in the progression of atherosclerotic vascular disease in diabetes, which is characterized by increased vascular smooth muscle cell (VSMC) proliferation and extracellular matrix accumulation. We previously reported that diabetes alters the activity of the kallikrein-kinin system and results in the upregulation of kinin receptors in the vessel wall. To determine whether glucose can directly influence the regulation of kinin receptors, the independent effect of high glucose (25 mM) on B(2)-kinin receptors (B2KR) in VSMC was examined. A threefold increase in B2KR protein levels and a 40% increase in B2KR surface receptors were observed after treatment with high glucose after 24 h. The mRNA levels of B2KR were also significantly increased by high glucose as early as 4 h later. To elucidate the cellular mechanisms by which glucose regulates B2KR, we examined the role of protein kinase C (PKC). High glucose increased total PKC activity and resulted in the translocation of conventional PKC isoforms (beta(1) and beta(2)), novel (epsilon), and atypical (zeta) PKC isoforms into the membrane. Inhibition of PKC activity prevented the increase in B2KR levels induced by ambient high glucose. These findings provide the first evidence that glucose regulates the expression of B(2) receptors in VSMC and provide a rationale to further study the interaction between glucose and kinins on the pathogenesis of atherosclerotic vascular disease in diabetes.


Subject(s)
Glucose/pharmacology , Muscle, Smooth, Vascular/physiology , Receptors, Bradykinin/genetics , Up-Regulation/drug effects , Animals , Aorta , Bradykinin/pharmacology , Calcium/metabolism , Cells, Cultured , Hyperglycemia , Isoenzymes/metabolism , Kinetics , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Protein Biosynthesis , Protein Kinase C/metabolism , Protein Kinase C beta , Protein Kinase C-epsilon , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptor, Bradykinin B2 , Transcription, Genetic
9.
J Pharmacol Exp Ther ; 296(1): 71-6, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11123364

ABSTRACT

Extensive research has provided few therapeutic agents for the treatment of septicemia. Bradykinin, an endogenous vasodepressor hormone, is a key mediator in the hypotension seen with septicemia. The present investigation shows that a stable metabolic fragment of bradykinin, arginine-proline-proline-glycine-phenylalanine (RPPGF), prevents the deleterious effects of endotoxin [lipopolysaccharide (LPS); a component of the membrane of Gram negative bacteria], the signaling agent responsible for the effects of septicemia, in both anesthetized rats and in isolated rat aortic segments. Survival time of rats treated with LPS (12 mg/kg) was significantly (p < 0.05) prolonged by pretreatment with RPPGF [140.3 +/- 16 min (n = 10)] compared with rats receiving saline and LPS [93.2 +/- 8 min (n = 39)]. Prolongation of survival was not seen when rats were pretreated with either bradykinin or with PRGFP (proline-arginine-glycine-phenylalanine-proline). Isolated aortic segments treated with LPS (30 microg/ml) showed a significantly reduced ability to contract in response to phenylephrine compared with control segments not receiving LPS. Pretreatment of the segments with RPPGF significantly reversed the LPS-induced reduction in contractile response of the segments. Removal of the endothelial layer did not alter the protection provided by RPPGF. These results demonstrate the ability of a stable metabolic fragment of bradykinin, RPPGF, to protect against the deleterious effects produced by LPS. The findings presented here may provide the basis for a new developmental area for novel therapeutic agents in the treatment of septicemia.


Subject(s)
Bradykinin/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Peptide Fragments/pharmacology , Shock, Septic/prevention & control , Animals , Aorta, Thoracic/drug effects , Blood Pressure/drug effects , Bradykinin/physiology , Heart Rate/drug effects , In Vitro Techniques , Lipopolysaccharides/toxicity , Male , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Peptide Fragments/physiology , Phenylephrine/pharmacology , Rats , Rats, Sprague-Dawley , Shock, Septic/mortality , Shock, Septic/physiopathology , Vasoconstrictor Agents/pharmacology
10.
Diabetes ; 49(12): 2160-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11118021

ABSTRACT

Glycation and/or oxidation of LDL may promote diabetic nephropathy. The mitogen-activated protein kinase (MAPK) cascade, which includes extracellular signal-regulated protein kinases (ERKs), modulates cell function. Therefore, we examined the effects of LDL on ERK phosphorylation in cultured rat mesangial cells. In cells exposed to 100 microg/ml native LDL or LDL modified by glycation, and/or mild or marked (copper-mediated) oxidation, ERK activation peaked at 5 min. Five minutes of exposure to 10-100 microg/ml native or modified LDL produced a concentration-dependent (up to sevenfold) increase in ERK activity. Also, 10 microg/ml native LDL and mildly modified LDL (glycated and/or mildly oxidized) produced significantly greater ERK activation than that induced by copper-oxidized LDL +/- glycation (P < 0.05). Pretreatment of cells with Src kinase and MAPK kinase inhibitors blocked ERK activation by 50-80% (P < 0.05). Native and mildly modified LDL, which are recognized by the native LDL receptor, induced a transient spike of intracellular calcium. Copper-oxidized (+/- glycation) LDL, recognized by the scavenger receptor, induced a sustained rise in intracellular calcium. The intracellular calcium chelator (EGTA/AM) further increased ERK activation by native and mildly modified LDL (P < 0.05). These findings demonstrate that native and modified LDL activate ERKs 1 and 2, an early mitogenic signal, in mesangial cells and provide evidence for a potential link between modified LDL and the development of glomerular injury in diabetes.


Subject(s)
Glomerular Mesangium/drug effects , Glomerular Mesangium/enzymology , Lipoproteins, LDL/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Animals , Calcium/physiology , Cells, Cultured , Enzyme Activation , Glomerular Mesangium/cytology , Glycosylation , Humans , Lipoproteins, LDL/metabolism , Male , Mitogen-Activated Protein Kinase Kinases/metabolism , Osmolar Concentration , Oxidation-Reduction , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Time Factors , src-Family Kinases/metabolism
11.
Am J Physiol Heart Circ Physiol ; 279(6): H2829-37, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11087238

ABSTRACT

Accumulation of extracellular matrix (ECM) is a hallmark feature of vascular disease. We have previously shown that hyperglycemia induces the expression of B(2)-kinin receptors in vascular smooth muscle cells (VSMC) and that bradykinin (BK) and hyperglycemia synergize to stimulate ECM production. The present study examined the cellular mechanisms through which BK contributes to VSMC fibrosis. VSMC treated with BK (10(-8) M) for 24 h significantly increased alpha(2)(I) collagen mRNA levels. In addition, BK produced a two- to threefold increase in alpha(2)(I) collagen promoter activity in VSMC transfected with a plasmid containing the alpha(2)(I) collagen promoter. Furthermore, treatment of VSMC with BK for 24 h produced a two- to threefold increase in the secretion rate of tissue inhibitor of metalloproteinase 1 (TIMP-1). The increase in alpha(2)(I) collagen mRNA levels and alpha(2)(I) collagen promoter activity, as well as TIMP-1 secretion, in response to BK were blocked by anti-transforming growth factor-beta (anti-TGF-beta) neutralizing antibodies. BK (10(-8) M) increased the endogenous production of TGF-beta1 mRNA and protein levels. Inhibition of the mitogen-activated protein kinase (MAPK) pathway by PD-98059 inhibited the increase of alpha(2)(I) collagen promoter activity, TIMP-1 production, and TGF-beta1 protein levels observed in response to BK. These findings provide the first evidence that BK induces collagen type I and TIMP-1 production via autocrine activation of TGF-beta1 and implicate MAPK pathway as a key player in VSMC fibrosis in response of BK.


Subject(s)
Bradykinin/pharmacology , MAP Kinase Signaling System/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Transforming Growth Factor beta/metabolism , Animals , Aorta/enzymology , Aorta/pathology , Collagen/genetics , Enzyme Inhibitors/pharmacology , Fibrosis , Flavonoids/pharmacology , Gene Expression/drug effects , MAP Kinase Signaling System/physiology , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/enzymology , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transcriptional Activation/drug effects , Transforming Growth Factor beta/genetics
12.
Hypertension ; 35(4): 942-7, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10775566

ABSTRACT

Bradykinin stimulates proliferation of aortic vascular smooth muscle cells (VSMCs). We investigated the action of bradykinin on the phosphorylation state of the mitogen-activated protein kinases p42(mapk) and p44(mapk) in VSMCs and tested the hypothesis that reactive oxygen species (ROS) might be involved in the signal transduction pathway linking bradykinin activation of nuclear transcription factors to the phosphorylation of p42(mapk) and p44(mapk). Bradykinin (10(-8) mol/L) rapidly increased (4- to 5-fold) the phosphorylation of p42(mapk) and p44(mapk) in VSMCs. Preincubation of VSMCs with either N-acetyl-L-cysteine and/or alpha-lipoic acid significantly decreased bradykinin-induced cytosolic and nuclear phosphorylation of p42(mapk) and p44(mapk). In addition, the induction c-fos mRNA levels by bradykinin was completely abolished by N-acetyl-L-cysteine and alpha-lipoic acid. Using the cell-permeable fluorescent dye dichlorofluorescein diacetate, we determined that bradykinin (10(-8) mol/L) rapidly increased the generation of ROS in VSMCs. The NADPH oxidase inhibitor diphenylene iodonium (DPI) blocked bradykinin-induced c-fos mRNA expression and p42(mapk) and p44(mapk) activation, implicating NADPH oxidase as the source for the generation of ROS. These findings demonstrate that the phosphorylation of cytosolic and nuclear p42(mapk) and p44(mapk) and the expression of c-fos mRNA in VSMCs in response to bradykinin are mediated via the generation of ROS and implicate ROS as important mediators in the signal transduction pathway through which bradykinin promotes VSMC proliferation in states of vascular injury.


Subject(s)
Bradykinin/pharmacology , MAP Kinase Signaling System/drug effects , Muscle, Smooth, Vascular/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Animals , Cell Division , Cells, Cultured , Enzyme Induction/drug effects , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Rats , Rats, Sprague-Dawley
13.
Am J Physiol ; 277(3): H1061-8, 1999 09.
Article in English | MEDLINE | ID: mdl-10484429

ABSTRACT

The vasoactive peptide bradykinin (BK) has been implicated in the pathophysiology of a number of vascular wall abnormalities, but the cellular mechanisms by which BK generates second messengers that alter vascular function are as yet undefined. Exposure of vascular smooth muscle cells (VSMC) to BK (10(-7) M) produced a rapid and transient rise in intracellular calcium, which preceded an increase in tyrosine phosphorylation of mitogen-activated protein kinase (MAPK). MAPK activation by BK was observed as early as 1 min, peaked at 5 min, and returned to baseline by 20 min. Treatment of cells with the intracellular calcium chelator EGTA-acetoxymethyl ester inhibited BK-stimulated MAPK activation, suggesting that intracellular calcium mobilization contributes to the activation of MAPK. The calmodulin inhibitor W-7 also markedly inhibited BK-induced MAPK phosphorylation in the cytoplasm as well as in the nucleus. Moreover, the BK-induced increase in c-fos mRNA levels was significantly inhibited by the calmodulin inhibitor, indicating that calmodulin is required for BK signaling leading to c-fos induction. These results implicate the calcium-calmodulin pathway in the mechanisms for regulating MAPK activity and the resultant c-fos expression induced by BK in VSMC.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calcium/metabolism , Calmodulin/metabolism , Muscle, Smooth, Vascular/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Signal Transduction , Animals , Bradykinin/pharmacology , Cells, Cultured , Male , Phosphorylation , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
14.
Am J Physiol ; 277(2): C253-61, 1999 08.
Article in English | MEDLINE | ID: mdl-10444401

ABSTRACT

Vascular smooth muscle cell (VSMC) proliferation is a prominent feature of the atherosclerotic process occurring after endothelial injury. A vascular wall kallikrein-kinin system has been described. The contribution of this system to vascular disease is undefined. In the present study we characterized the signal transduction pathway leading to mitogen-activated protein kinase (MAPK) activation in response to bradykinin (BK) in VSMC. Addition of 10(-10)-10(-7) M BK to VSMC resulted in a rapid and concentration-dependent increase in tyrosine phosphorylation of several 144- to 40-kDa proteins. This effect of BK was abolished by the B(2)-kinin receptor antagonist HOE-140, but not by the B(1)-kinin receptor antagonist des-Arg(9)-Leu(8)-BK. Immunoprecipitation with anti-phosphotyrosine antibodies followed by immunoblot revealed that 10(-9) M BK induced tyrosine phosphorylation of focal adhesion kinase (p125(FAK)). BK (10(-8) M) promoted the association of p60(src) with the adapter protein growth factor receptor binding protein-2 and also induced a significant increase in MAPK activity. Pertussis and cholera toxins did not inhibit BK-induced MAPK tyrosine phosphorylation. Protein kinase C downregulation by phorbol 12-myristate 13-acetate and/or inhibitors to protein kinase C, p60(src) kinase, and MAPK kinase inhibited BK-induced MAPK tyrosine phosphorylation. These findings provide evidence that activation of the B(2)-kinin receptor in VSMC leads to generation of multiple second messengers that converge to activate MAPK. The activation of this crucial kinase by BK provides a strong rationale to investigate the mitogenic actions of BK on VSMC proliferation in disease states of vascular injury.


Subject(s)
Bradykinin/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Animals , Cells, Cultured , Cholera Toxin/pharmacology , Cytoplasm/enzymology , Enzyme Activation/physiology , Male , Muscle, Smooth, Vascular/cytology , Phosphorylation/drug effects , Protein Kinase C/physiology , Protein-Tyrosine Kinases/drug effects , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Bradykinin/physiology , Tyrosine/metabolism , Virulence Factors, Bordetella/pharmacology
15.
Biol Chem ; 379(10): 1271-7, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9820588

ABSTRACT

Aprotinin, an inhibitor of the enzymatic activity of kallikrein in vitro, has been used to study the possible contributions of the kallikrein-kinin systems to physiological and pathological conditions. Pharmacokinetic studies indicate that aprotinin is concentrated in the kidney; however, there is little information with regard to its cellular distribution. The purpose of the present work was to study the cellular distribution of aprotinin, which would be valuable for a better understanding of its intrarenal effects. Sprague-Dawley rats (200-250g, n = 36) received aprotinin (50000 KIU/rat) and were killed at different intervals after its administration. The kidneys were examined histologically and the cellular distribution of aprotinin was studied by immunohistochemistry. Aprotinin was localized at 30 min concentrated within vesicles in the apical border of the proximal tubule cells. Later (2 h) it was observed distributed over the cytoplasm, where it remained for the 24 h studied. Aprotinin was also detected in connecting tubule cells colocalized with kallikrein, and in the basal portion of collecting tubule cells. No evidence of endogenous aprotinin was observed. The binding of aprotinin to the connecting tubule cells and collecting ducts offers a partial explanation of its renal effects.


Subject(s)
Aprotinin/pharmacokinetics , Kidney/metabolism , Serine Proteinase Inhibitors/pharmacokinetics , Animals , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley
16.
Hypertension ; 31(4): 978-85, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9535424

ABSTRACT

Oleic acid and angiotensin II (Ang II) are elevated and may interact to accelerate vascular disease in obese hypertensive patients. We studied the effects of oleic acid and Ang II on growth responses of rat aortic smooth muscle cells (VSMCs). Oleic acid (50 micromol/L) raised thymidine incorporation by 50% at 24 hours and cell number by 55% at 6 days (P<.05). Ang II (10(-11) to 10(-6) mol/L) did not significantly increase thymidine incorporation or VSMC number. Combining Ang II and 50 micromol/L oleic acid doubled thymidine incorporation and VSMC number. Losartan, an angiotensin type 1 (AT1) receptor antagonist, blocked the synergistic interaction between Ang II and oleic acid, whereas the AT2 receptor antagonist PD 123319 did not. Protein kinase C inhibition and downregulation, as well as inhibition of extracellular signal-regulated kinase (ERK) activation by PD 98059, eliminated the rise of thymidine incorporation in response to oleic acid and the synergistic interaction with Ang II. However, the response to 10% fetal bovine serum was unaffected. An antisense oligodeoxynucleotide to ERK-1 and ERK-2 reduced ERK protein expression and activation by 83% and 75%, respectively. Antisense prevented the rise of thymidine incorporation in response to oleic acid and the synergy with Ang II. Antisense reduced but did not prevent increased thymidine incorporation in response to serum. The data indicate that oleic acid and Ang II exert a synergistic mitogenic effect in VSMCs and suggest an important role for the AT1 receptor, PKC, and ERK in this synergy. The observations raise the possibility that a synergistic mitogenic interaction between oleic acid and Ang II accelerates vascular remodeling in obese hypertensive patients.


Subject(s)
Angiotensin II/pharmacology , Mitogen-Activated Protein Kinases , Muscle, Smooth, Vascular/drug effects , Oleic Acid/pharmacology , Pharmaceutic Aids/pharmacology , Vasoconstrictor Agents/pharmacology , Angiotensin Receptor Antagonists , Animals , Aorta/cytology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Cell Count/drug effects , Cell Division/drug effects , Cells, Cultured/drug effects , Drug Synergism , JNK Mitogen-Activated Protein Kinases , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Oligonucleotides, Antisense/pharmacology , Rats , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , p38 Mitogen-Activated Protein Kinases
17.
Diabetes ; 46(12): 2049-56, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9392495

ABSTRACT

The renal kallikrein-kinin system and the renin-angiotensin system are implicated in the pathogenesis of diabetic nephropathy. We have shown that renal kallikrein and renin gene expression are altered by diabetes. To investigate the cellular mechanisms responsible for these changes, we examined the effects of acute insulin and insulin-like growth factor I (IGF-I) treatment on renal kallikrein-kinin and renin-angiotensin system components. Three weeks after induction of diabetes, we measured renal kallikrein and renin mRNA levels, renal kallikrein and renal renin activity, and plasma renin activity in control and diabetic rats and diabetic rats treated with insulin or IGF-I for 2 or 5 h. In diabetic rats, kallikrein and renin mRNA levels were reduced >50% compared with control rats. Renal tissue kallikrein levels and plasma renin activity were decreased, whereas renal renin content was unchanged. Insulin increased kallikrein and renin mRNA levels after 2 h. IGF-I, at a dosage that stimulated kallikrein mRNA levels in control rats, had no effect on renal kallikrein and renin content or mRNA levels in diabetic rats. However, infusion of a fivefold higher IGF-I dosage resulted in a two- to threefold increase in kallikrein and renin mRNA levels in 2 h. These data suggest that 1) diabetes suppresses kallikrein and renin gene expression, and these abnormalities are reversed by insulin or IGF-I; and 2) the diabetic state produces resistance to IGF-I induction of kallikrein and renin gene expression. These changes in regulated synthesis of kallikrein and renin in the kidney may underlie renal vascular changes that develop in diabetes.


Subject(s)
Diabetes Mellitus, Experimental/enzymology , Gene Expression/drug effects , Insulin-Like Growth Factor I/pharmacology , Insulin/pharmacology , Kallikreins/genetics , Kidney/enzymology , Renin/genetics , Animals , Immunohistochemistry , Kallikreins/analysis , Kallikreins/metabolism , Male , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Renin/analysis , Renin/metabolism
18.
Biochem Pharmacol ; 53(12): 1823-32, 1997 Jun 15.
Article in English | MEDLINE | ID: mdl-9256157

ABSTRACT

Thromboxane A2 (TXA2) interacts with its G-protein coupled receptor, the TP receptor, to produce contraction and proliferation of vascular smooth muscle cells. We have shown previously that proliferation of primary cultures of vascular smooth muscle cells initiated by [1S-(1alpha, 2beta(5Z), 3alpha(1E, 3R), 4alpha]-7-[3-(3-hydroxy-4-(4'-iodophenoxy)-1-butenyl)-7-oxab icyclo-[2.2.1]heptan-2yl]-5'-heptenoic acid (I-BOP), a stable TXA2 mimetic, is mediated by activation of mitogen-activated protein (MAP) kinase. In the present study, we examined further the intracellular mediators involved in TXA2 activation of vascular smooth muscle cells. Transient transfection of the cDNA for the TP receptor into A7r5 vascular smooth muscle cells resulted in expression of TP receptors with a receptor density, Bmax, of 0.7 +/- 0.2 pmol/mg protein and a receptor affinity, Kd, of 0.6 +/- 0.1 nM (N = 7). Mock transfected cells lacked significant receptor expression. In TP receptor transfected cells, I-BOP increased the activation of MAP kinase 2-fold, stimulated tyrosine phosphorylation of cellular proteins of relative molecular mass (Mr) of 140, 85, 60, 56, and 45 kDa, and increased the message for c-jun, a nuclear transcription factor involved in mitogenesis, 2.6-fold. Immunoblot analysis indicated that the 85-kDa protein represented phosphoinositide 3-kinase (PI3-K), while the 60 kDa protein was the TP receptor. The activity of PI3-K was increased 3.5-fold by the addition of I-BOP (0.1 microM). In summary, the present study demonstrated that stimulation of the TP receptor results in tyrosine phosphorylation of the receptor and of PI3-K.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Fatty Acids, Unsaturated/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, Thromboxane/metabolism , Tyrosine/metabolism , Cell Division , Cell Line , Enzyme Activation , Genes, jun , Muscle, Smooth, Vascular/drug effects , Phosphatidylinositol 3-Kinases , Phosphorylation/drug effects , Phosphotransferases (Alcohol Group Acceptor)/chemistry , RNA, Messenger/biosynthesis , Radioligand Assay , Signal Transduction , Transfection
19.
Hypertension ; 29(3): 757-62, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9052892

ABSTRACT

Bradykinin and lys-bradykinin generated intrarenally appear to be important renal paracrine hormones. However, the renal effects of endogenously generated bradykinin are still not clearly defined. In this study, we measured acute changes in renal excretory and hemodynamic functions and renal cortical interstitial fluid levels of bradykinin, prostaglandin E2, and cGMP in response to an acute intrarenal arterial infusion of the bradykinin B2 receptor antagonist Hoe 140 (icatibant), cyclooxygenase inhibitor indomethacin, or nitric oxide synthase inhibitor N(G)-monomethyl-L-arginine (L-NMMA) given individually or combined in uninephrectomized, conscious dogs (n=10) in low sodium balance. Icatibant caused a significant decrease in urine flow, urinary sodium excretion, and renal plasma flow rate (each P<.001). Glomerular filtration rate did not change during icatibant administration. Icatibant produced an unexpected large increase in renal interstitial fluid bradykinin (P<.0001) while decreasing renal interstitial fluid prostaglandin E2 and cGMP (each P<.001). Both indomethacin and L-NMMA when given individually caused significant antidiuresis and antinatriuresis and decreased renal blood flow (each P<.001). Glomerular filtration rate decreased during L-NMMA administration (P<.001) and did not change during indomethacin administration. Combined administration of icatibant and indomethacin or L-NMMA caused significant decreases in renal excretory and hemodynamic functions, which were not different from changes observed with icatibant alone. The failure of icatibant to change renal function after inhibition of cyclooxygenase and nitric oxide synthase activity suggests that the effects of kinin B2 receptor are mediated by intrarenal prostaglandin E2 and nitric oxide generation. The increase in renal interstitial fluid bradykinin during icatibant requires further study of possible alterations in kinin synthesis, degradation, or clearance as a result of B2 receptor blockade.


Subject(s)
Bradykinin Receptor Antagonists , Bradykinin/analogs & derivatives , Bradykinin/physiology , Dinoprostone/metabolism , Kidney/drug effects , Nitric Oxide/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bradykinin/pharmacology , Dogs , Drug Interactions , Enzyme Inhibitors/pharmacology , Female , Hemodynamics/drug effects , Indomethacin/pharmacology , Kidney/metabolism , Renal Circulation/drug effects , omega-N-Methylarginine/pharmacology
20.
Am J Physiol ; 273(6): F916-24, 1997 12.
Article in English | MEDLINE | ID: mdl-9435680

ABSTRACT

Glomerular hypertension and glomerular hypertrophy act early and synergistically to promote glomerular injury in diabetes. We have previously shown that increased renal kinin production contributes to the glomerular hemodynamic abnormalities associated with diabetes. Glomerulosclerosis, characterized by mesangial cell proliferation and matrix expansion, is the final pathway leading to renal failure. The signal(s) initiating mesangial cell proliferation is ill defined. In the present study, we utilized immunofluorescence, immunoprecipitation, and immunoblotting techniques to identify substrates that are tyrosine phosphorylated in response to bradykinin action in mesangial cells. Immunofluorescence microscopy of mesangial cells stained with anti-phosphotyrosine (anti-PY) antibodies following bradykinin treatment (10(-9)-10(-6) M) revealed a dose-dependent increase in the labeling of cytoplasmic and nuclear proteins. Immunoprecipitation with anti-PY, followed by immunoblot revealed bradykinin-induced tyrosyl phosphorylation of tubulin and mitogen-activated protein kinase (MAPK). Confocal microscopy of mesangial cells stained for MAPK indicated that bradykinin stimulation resulted in translocation of MAPK from the cytoplasm to the nucleus by 2 h. These data demonstrate that bradykinin action results in the tyrosine phosphorylation of cellular proteins in mesangial cells and suggest a role for tubulin and MAPK in the signaling cascade of bradykinin leading to altered mesangial function.


Subject(s)
Bradykinin/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Nucleus/enzymology , Glomerular Mesangium/metabolism , Tubulin/metabolism , Animals , Cells, Cultured , Cytosol/enzymology , Glomerular Mesangium/cytology , Glomerular Mesangium/drug effects , Kinetics , Male , Microscopy, Confocal , Phosphorylation , Phosphotyrosine/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Tubulin/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...