Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Lung Cancer ; 58(3): 422-4, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17643548

ABSTRACT

Gefitinib is mainly metabolized by the liver and its excretion is mostly in bile excrements. However, the feasibility of gefitinib in patients with chronic renal failure undergoing hemodialysis has not, so far, been reported. A 58-year-old woman with chronic renal failure due to polycystic kidney disease, undergoing hemodialysis, experienced diplopia due to meningitis carcinomatosa by lung adenocarcinoma. Sequencing analysis of her tumor tissue revealed deletion of 15 nucleotides in E746-A750 of exon 19. She started daily administration of 250mg gefitinib with hemodialysis three times a week. Her pharmacokinetic pattern after gefitinib administration was similar to those in patients with normal renal function and 88.7% of gefitinib was kept in the plasma through hemodialysis. Her symptoms and signs of meningitis carcinomatosa on brain magnetic resonance images improved. Thirteen months later, the meningitis got worse again and she stopped gefitinib administration. During gefitinib administration, there were no signs of adverse events. In summary, gefitinib is not eliminated by hemodialysis and was safely administered to a patient with non-small cell lung cancer and chronic renal failure who was undergoing hemodialysis.


Subject(s)
Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Quinazolines/therapeutic use , Renal Dialysis , Female , Gefitinib , Humans , Magnetic Resonance Imaging , Middle Aged , Quinazolines/administration & dosage , Quinazolines/pharmacokinetics
2.
Biol Chem ; 387(2): 203-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16497153

ABSTRACT

An angiotensin-converting enzyme inhibitor (ACE-I) reduces cardiac remodeling and a bradykinin B2 receptor (B2R) antagonist partially abolishes this ACE-I effect. However, bradykinin has two different types of receptor, the B1 receptor (B1R) and B2R. Although B1R is induced under several pathological conditions, including hypertension, the role of cardiac B1R in hypertension is not clear. We therefore investigated the role of cardiac B1R in stroke-prone spontaneously hypertensive rats (SHR-SP) and Wistar-Kyoto (WKY) rats. The B1R mRNA expression level in the heart was significantly higher in SHR-SP than in WKY rats. Chronic infusion of a B1R antagonist for 4 weeks significantly elevated blood pressure and left-ventricular weight of SHR-SP. Morphological analysis indicated that cardiomyocyte size and cardiac fibrosis significantly increased after administration of the B1R antagonist. The phosphorylation of mitogen-activated protein (MAP) kinases, including ERK, p38, and JNK, was significantly increased in the hearts of SHR-SP rats receiving the B1R antagonist. The TGF-beta1 expression level was significantly increased in SHR-SP rats treated with the B1R antagonist compared to that in WKY rats. The B1R antagonist significantly increased phosphorylation of Thr495 in endothelial nitric oxide synthase (eNOS), which is an inhibitory site of eNOS. These results suggest that the role of B1R in the heart may be attenuation of cardiac remodeling via inhibition of the expression of MAP kinases and TGF-beta1 through an increase in eNOS activity in a hypertensive condition.


Subject(s)
Disease Models, Animal , Heart/physiopathology , Hypertension/physiopathology , Receptor, Bradykinin B1/metabolism , Stroke/physiopathology , Ventricular Remodeling , Animals , Antihypertensive Agents/therapeutic use , Bradykinin B1 Receptor Antagonists , Enzyme Activation/drug effects , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Heart/drug effects , Hypertension/drug therapy , Male , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Nitric Oxide Synthase Type III/drug effects , Nitric Oxide Synthase Type III/metabolism , RNA, Messenger/drug effects , RNA, Messenger/genetics , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptor, Bradykinin B1/genetics , Stroke/drug therapy , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta1 , Ventricular Remodeling/drug effects , Ventricular Remodeling/physiology
3.
Hypertens Res ; 29(11): 865-74, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17345786

ABSTRACT

Angiotensin II receptor blockers (ARBs) are widely used for the treatment of hypertension. It is believed that treatment with an ARB increases the level of plasma angiotensin II (Ang II) because of a lack of negative feedback on renin activity. However, Ichikawa (Hypertens Res 2001; 24: 641-646) reported that long-term treatment of hypertensive patients with olmesartan resulted in a reduction in plasma Ang II level, though the mechanism was not determined. It has been reported that angiotensin 1-7 (Ang-(1-7)) potentiates the effect of bradykinin and acts as an angiotensin-converting enzyme (ACE) inhibitor. It is known that ACE2, which was discovered as a novel ACE-related carboxypeptidase in 2000, hydrolyzes Ang I to Ang-(1-9) and also Ang II to Ang-(1-7). It has recently been reported that olmesartan increases plasma Ang-(1-7) through an increase in ACE2 expression in rats with myocardial infarction. We hypothesized that over-expression of ACE2 may be related to a reduction in Ang II level and the cardioprotective effect of olmesartan. Administration of 0.5 mg/kg/day of olmesartan for 4 weeks to 12-week-old stroke-prone spontaneously hypertensive rats (SHRSP) significantly reduced blood pressure and left ventricular weight compared to those in SHRSP given a vehicle. Co-administration of olmesartan and (D-Ala7)-Ang-(1-7), a selective Ang-(1-7) antagonist, partially inhibited the effect of olmesartan on blood pressure and left ventricular weight. Interestingly, co-administration of (D-Ala7)-Ang-(1-7) with olmesartan significantly increased the plasma Ang II level (453.2+/-113.8 pg/ml) compared to olmesartan alone (144.9+/-27.0 pg/ml, p<0.05). Moreover, olmesartan significantly increased the cardiac ACE2 expression level compared to that in Wistar Kyoto rats and SHRSP treated with a vehicle. Olmesartan significantly improved cardiovascular remodeling and cardiac nitrite/ nitrate content, but co-administration of olmesartan and (D-Ala7)-Ang-(1-7) partially reversed this anti-remodeling effect and the increase in nitrite/nitrate. These findings suggest that olmesartan may exhibit an ACE inhibitory action in addition to an Ang II receptor blocking action, prevent an increase in Ang II level, and protect cardiovascular remodeling through an increase in cardiac nitric oxide production and endogenous Ang-(1-7) via over-expression of ACE2.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II/drug effects , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Peptide Fragments/drug effects , Peptidyl-Dipeptidase A/drug effects , Tetrazoles/pharmacology , Aldosterone/blood , Angiotensin I , Angiotensin-Converting Enzyme 2 , Animals , Blood Pressure/drug effects , Gene Expression , Male , Myocardium/metabolism , Nitrates/metabolism , Nitrites/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Renin/blood , Ventricular Remodeling/drug effects
4.
Diabetes ; 54(5): 1573-80, 2005 May.
Article in English | MEDLINE | ID: mdl-15855348

ABSTRACT

We investigated the role of the kallikrein-kinin system in cardiac function and glucose utilization in the streptozotocin (STZ)-induced diabetic rat model using a gene transfer approach. Adenovirus harboring the human tissue kallikrein gene was administered to rats by intravenous injection at 1 week after STZ treatment. Human kallikrein transgene expression was detected in the serum and urine of STZ-induced diabetic rats after gene transfer. Kallikrein gene delivery significantly reduced blood glucose levels and cardiac glycogen accumulation in STZ-induced diabetic rats. Kallikrein gene transfer also significantly attenuated elevated plasma triglyceride and cholesterol levels, food and water intake, and loss of body weight gain, epididymal fat pad, and gastrocnemius muscle weight in STZ-induced diabetic rats. However, these effects were blocked by icatibant, a kinin B2 receptor antagonist. Cardiac function was significantly improved after kallikrein gene transfer as evidenced by increased cardiac output and +/-delta P/delta t (maximum speed of contraction/relaxation), along with elevated cardiac sarco(endo)plasmic reticulum (Ca2+ + Mg2+)-ATPase (SERCA)-2a, phosphorylated phospholamban, NOx and cAMP levels, and GLUT4 translocation into plasma membranes of cardiac and skeletal muscle. Kallikrein gene delivery also increased Akt and glycogen synthase kinase (GSK)-3beta phosphorylation, resulting in decreased GSK-3beta activity in the heart. These results indicate that kallikrein through kinin formation protects against diabetic cardiomyopathy by improving cardiac function and promoting glucose utilization and lipid metabolism.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Experimental/physiopathology , Kallikreins/genetics , Lipids/blood , Adenoviridae , Animals , Blood Glucose/drug effects , Cardiomyopathies/prevention & control , Diabetes Mellitus, Experimental/blood , Disease Models, Animal , Genetic Vectors , Glycogen/metabolism , Humans , Kallikreins/pharmacology , Male , Myocardium/metabolism , Myocardium/pathology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Transfection/methods
5.
Life Sci ; 76(21): 2457-71, 2005 Apr 08.
Article in English | MEDLINE | ID: mdl-15763077

ABSTRACT

Nitric oxide (NO) has been shown to play a key role in the regulation of cardiac hypertrophy and fibrosis in response to myocardial ischemia in part by antagonizing the action of angiotensin II (Ang II). In this study, we investigated the potential protective role of human endothelial nitric oxide synthase (eNOS) in left ventricular (LV) remodeling after myocardial infarction (MI) by a somatic gene transfer approach. Male Wistar rats underwent coronary artery ligation to induce MI. One week after surgery, adenovirus encoding the human eNOS or luciferase gene under the control of the CMV promoter/enhancer was injected into rats via the tail vein, and animals were sacrificed at 1 and 5 weeks after gene transfer. Successful gene transfer was evaluated based on increased levels of NO and cGMP in the heart, measured at one week after eNOS gene delivery. Six weeks after MI, the LV end-diastolic pressure, heart weight, LV axis length and cardiomyocyte size were markedly increased compared to the Sham group, while eNOS gene delivery significantly reduced these parameters. Rats receiving control virus developed considerably more fibrotic lesions identified by Sirius Red staining and collagen I immunostaining compared to Sham rats, and eNOS gene delivery significantly reduced collagen accumulation. eNOS gene transfer also reduced TUNEL-positive apoptotic cells. The cardioprotective effect of NO was accompanied by reduced NADH and NADPH oxidase activities and superoxide formation, TGF-beta1 and p27 levels, JNK activation, NF-kappa B nuclear translocation, and caspase-3 activity. This study shows that NO may play an important role in attenuating cardiac remodeling and apoptosis after myocardial infarction via suppression of oxidative stress-mediated signaling pathways.


Subject(s)
Genetic Therapy/methods , Myocardial Infarction/therapy , Nitric Oxide Synthase/genetics , Oxidative Stress/genetics , Ventricular Remodeling/genetics , Adenoviridae , Animals , Apoptosis/physiology , Azo Compounds , Caspase 3 , Caspases/metabolism , Cell Cycle Proteins/metabolism , Collagen , Coronary Vessels/surgery , Cyclic GMP/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Genetic Vectors , Heart Ventricles/pathology , Humans , Immunoblotting , Immunohistochemistry , In Situ Nick-End Labeling , JNK Mitogen-Activated Protein Kinases/metabolism , Ligation , Luciferases/genetics , MAP Kinase Kinase 4 , Male , Mitogen-Activated Protein Kinase Kinases/metabolism , NAD/metabolism , NADP/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III , Organ Size , Rats , Rats, Wistar , Superoxides/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , Tumor Suppressor Proteins/metabolism , Ventricular Function
6.
Circ J ; 68(12): 1194-8, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15564706

ABSTRACT

BACKGROUND: High pulse wave velocity (PWV) is related to cardiovascular risk in essential hypertension (EHT). It is reported that short-term treatment with an angiotensin II receptor blocker (ARB) decreases PWV, as well as blood pressure (BP), and increases the serum adiponectin, known as an adipocytokine, which has an anti-atherosclerotic effect. However, it is not known whether long-term treatment with ARB prevents the increase in PWV independently of the reduction of BP, and whether adiponectin is related to the chronic effect of ARB on PWV. METHODS AND RESULTS: In order to examine the short-term effect of ARB on PWV, 9 subjects with EHT had PWV measured before and after treatment with an ARB for 1 month. The treatment significantly reduced PWV and BP. For evaluation of the long-term effect of ARB therapy, 56 consecutive subjects with EHT who were already taking anti-hypertensive drugs other than an angiotensin-converting enzyme inhibitor had their PWV measured. We divided the EHT subjects into 2 groups: (1) the ARB group (EHT treated with an ARB for at least 6 months) and (2) the control group (EHT treated with anti-hypertensive drugs other than an ARB). Although there was no significant difference between the 2 groups in BP, age or body mass index, the PWV value in the ARB group was significantly lower than that in the control group. Moreover, the serum adiponectin concentration in the ARB group was significantly higher than that in the control group. CONCLUSIONS: Long-term treatment with ARB inhibits the progression of arterial stiffness independent of BP reduction. One of the mechanisms may be related to the increased serum adiponectin concentration after treatment with an ARB.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Arteries/drug effects , Arteries/physiopathology , Hypertension/drug therapy , Hypertension/physiopathology , Adiponectin , Adrenergic Antagonists/therapeutic use , Antihypertensive Agents/therapeutic use , Blood Flow Velocity/drug effects , Blood Pressure/drug effects , Calcium Channel Blockers/therapeutic use , Diuretics/therapeutic use , Drug Therapy, Combination , Elasticity , Female , Humans , Intercellular Signaling Peptides and Proteins/blood , Male , Middle Aged , Osmolar Concentration , Pulse
7.
Hypertens Res ; 27(6): 399-408, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15253105

ABSTRACT

The kallikrein-kinin system plays important roles in blood pressure regulation, metabolism of electrolytes and organ protection. Although the bradykinin B2 receptor (B2R) has been reported to be involved in most of these effects, a role of the bradykinin B1 receptor (B1R) has also been noted recently. The aim of this study was to determine the role of renal B1R in stroke-prone spontaneously hypertensive rats (SHR-SP). Sixteen-week-old SHR-SP and Wistar Kyoto rats (WKY) as a control were used in the experiments. A high level of B1R mRNA was detected in SHR-SP, while the expression in WKY was almost undetectable. Immunohistochemistry revealed a B1R protein in the renal tubules and glomeruli in SHR-SP. The acute injection of a B1 R agonist into SHR-SP increased urinary NOx excretion to a level up to 5-fold higher than that in the SHR-SP treated with vehicle. The infusion of B1 R antagonist for 4 weeks resulted in a significant elevation of blood pressure and urinary albumin excretion and a decrease in urinary NOx excretion in SHR-SP. The administration of B1 R antagonist resulted in renal interstitial and glomerular fibrosis in SHR-SP. Moreover, the expressions of transforming growth factor (TGF) beta1 protein and collagen III mRNA in SHR-SP treated with B1R antagonist were significantly higher than those of SHR-SP treated with a vehicle. The expression and phosphorylation of extracellular signal-regulated protein kinase (ERK) and p38, but not c-Jun N-terminal kinase (JNK), were significantly increased in the SHR-SP treated with B1R antagonist. These results indicated that renal B1R might be over-expressed in a high blood pressure condition, and that this upregulated B1 R may play an important role in renal protection by inhibiting renal fibrosis via an increase of NO production and a suppression of TGFbeta1 expression and mitogen-activated protein kinase (ERK and p38) phosphorylation.


Subject(s)
Hypertension, Renal/physiopathology , Kidney Cortex/physiology , Receptor, Bradykinin B1/genetics , Stroke/physiopathology , Animals , Blood Pressure/physiology , Bradykinin B1 Receptor Antagonists , Hypertension, Renal/genetics , Kallikrein-Kinin System/physiology , Male , Nitrogen Compounds/urine , RNA, Messenger/analysis , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptor, Bradykinin B1/agonists , Stroke/genetics
8.
Am J Physiol Heart Circ Physiol ; 285(4): H1506-14, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12805025

ABSTRACT

Adrenomedullin (AM) has been shown to protect against cardiac remodeling. In this study, we investigated the potential role of AM in myocardial ischemia-reperfusion (I/R) injury through adenovirus-mediated gene delivery. One week after AM gene delivery, rats were subjected to 30-min coronary occlusion, followed by 2-h reperfusion. AM gene transfer significantly reduced the ratio of infarct size to ischemic area at risk and the occurrence of sustained ventricular fibrillation compared with control rats. AM gene delivery also attenuated apoptosis, assessed by both terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assay and DNA laddering. The effect of AM gene transfer on infarct size, arrhythmia, and apoptosis was abolished by an AM antagonist, calcitonin gene-related peptide [CGRP(8-37)]. Expression of human AM significantly increased cardiac cGMP levels and reduced superoxide production, superoxide density, NAD(P)H oxidase activity, p38 MAPK activation, and Bax levels. Moreover, AM increased Akt and Bad phosphorylation and Bcl-2 levels, but decreased caspase-3 activation. These results indicate that AM protects against myocardial infarction, arrhythmia, and apoptosis in I/R injury via suppression of oxidative stress-induced Bax and p38 MAPK phosphorylation and activation of the Akt-Bad-Bcl-2 signaling pathway. Successful application of this technology may have a protective effect in coronary artery diseases.


Subject(s)
Apoptosis , Gene Transfer Techniques , Myocardial Infarction/prevention & control , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/physiopathology , Peptides/genetics , Adrenomedullin , Animals , Arrhythmias, Cardiac/prevention & control , Blotting, Western , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Hemodynamics , Humans , Male , Myocardial Infarction/pathology , Myocardium/metabolism , Myocytes, Cardiac , NADPH Oxidases/antagonists & inhibitors , Peptides/metabolism , Proteins/metabolism , Rats , Rats, Wistar , Superoxides/antagonists & inhibitors
9.
Regul Pept ; 112(1-3): 115-20, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12667632

ABSTRACT

Adrenomedullin (AM) is a potent vasodilator expressed in tissues relevant to cardiovascular function. AM peptide has been shown to inhibit the proliferation and migration of vascular smooth muscle cells in vitro. However, the effect of AM on blood vessels after vascular injury in vivo has not been elucidated. In order to explore the potential roles of AM in vascular biology, we evaluated the effect of AM by local gene delivery on neointima formation in balloon-injured rat artery. Adenovirus carrying the human AM cDNA under the control of cytomegalovirus promoter/enhancer (Ad.CMV-hAM) was generated by homologous recombination. After delivery of Ad.CMV-hAM into rat left carotid artery, we identified the expression of human AM mRNA in the left carotid artery, but not in the right carotid artery, heart or kidney by reverse transcription-polymerase chain reaction (RT-PCR) followed by Southern blot analysis. Following local AM gene delivery, we observed a 51% reduction in intima/media ratio at the injured site as compared with that of control rats injected with the luciferase gene (n=7, P<0.01). AM gene transfer resulted in regeneration of endothelium as compared to the control. AM gene delivery significantly increased cGMP levels in balloon-injured arteries. These results indicate that AM contributes to reduction of neointima formation by promotion of re-endothelialization and inhibition of vascular smooth muscle cell proliferation via cGMP-dependent signaling pathway.


Subject(s)
Adenoviridae/genetics , Angioplasty, Balloon , Arterial Occlusive Diseases/therapy , Genetic Vectors , Peptides/genetics , Adrenomedullin , Animals , Arterial Occlusive Diseases/etiology , Arterial Occlusive Diseases/pathology , Carotid Arteries/pathology , Carotid Stenosis/etiology , Carotid Stenosis/pathology , Carotid Stenosis/therapy , Cyclic AMP , Cyclic GMP , Gene Expression , Humans , Models, Biological , Peptides/metabolism , RNA, Messenger/metabolism , Rats , Tunica Intima/pathology
10.
J Biol Chem ; 278(1): 219-25, 2003 Jan 03.
Article in English | MEDLINE | ID: mdl-12411434

ABSTRACT

We demonstrated that rat kinin B(1) receptors displayed a ligand-independent constitutive activity, assessed through inositol phosphate production in transiently or stably transfected human embryonic kidney 293A cells. Substitution of Ala for Asn(130) in the third transmembrane domain resulted in additional constitutive activation of the B(1) receptor. The constitutively active mutant N130A receptor could be further activated by the B(1) receptor agonist des-Arg(9)-bradykinin. To gain insights into the physiological function of the B(1) receptor, we have generated transgenic mice overexpressing wild-type and constitutively active mutant receptors under the control of human cytomegalovirus immediately early gene enhancer/promoter. The rat B(1) receptor transgene expression was detected in the aorta, brain, heart, lung, liver, kidney, uterus, and prostate of transgenic mice by reverse transcription-polymerase chain reaction/Southern blot analysis. Transgenic mice were fertile and normotensive. Overexpression of B(1) receptors exacerbated paw edema induced by carrageenan and rendered transgenic mice more susceptible to lipopolysaccharide-induced endotoxic shock. Interestingly, the hemodynamic response to kinins was altered in transgenic mice, with des-Arg(9)-bradykinin inducing blood pressure increase when intravenously administered. Our study supports an important role for B(1) receptors in modulating inflammatory responses and for the first time demonstrates that B(1) receptors mediate a hypertensive response to des-Arg(9)-bradykinin.


Subject(s)
Blood Pressure/physiology , Bradykinin/analogs & derivatives , Bradykinin/metabolism , Inflammation/metabolism , Receptors, Bradykinin/metabolism , Amino Acid Sequence , Animals , Blood Pressure/drug effects , Bradykinin/pharmacology , Carrageenan/toxicity , Cell Line , Female , Humans , Inflammation/chemically induced , Inositol Phosphates/metabolism , Ligands , Lipopolysaccharides/pharmacology , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Promoter Regions, Genetic , Protein Structure, Secondary , Rats , Receptor, Bradykinin B1 , Receptors, Bradykinin/genetics , Second Messenger Systems/physiology , Survival Rate , Transgenes
11.
Hypertension ; 40(5): 653-9, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12411458

ABSTRACT

In this study, we used the somatic gene delivery approach to explore the role of the kallikrein-kinin system (KKS) in cardiac remodeling and apoptosis after myocardial infarction (MI). Rats were subjected to coronary artery ligation to induce MI, and adenovirus carrying the human tissue kallikrein or luciferase gene was injected into the tail vein at 1 week after surgery. Cardiac output gradually decreased from 2 to 6 weeks after MI, whereas delivery of the kallikrein gene prevented this decrease. Cardiac responses to dobutamine-induced stress were improved in rats receiving kallikrein gene as compared with rats receiving control virus at 6 weeks after MI. Kallikrein significantly improved cardiac remodeling by decreasing collagen density, cardiomyocyte size, and left ventricular internal perimeter and increasing capillary density in the heart at 6 weeks after MI. Kallikrein gene transfer attenuated myocardial apoptosis, which was positively correlated with remodeling parameters in the heart at 2 weeks after MI. Endothelial dysfunction, characterized by increased vascular resistance, decreased left ventricular blood flow, and decreased cardiac nitric oxide levels, existed in remodeled hearts at 2 weeks after MI, whereas kallikrein gene transfer improved these parameters. Kallikrein gene delivery improved cell survival parameters as shown by increased phospho-Akt and reduced caspase-3 activation at 2 weeks after MI. This study indicates that the kallikrein-kinin system plays an important role in preventing the progression of heart failure by attenuating cardiac hypertrophy and fibrosis, improving endothelial function, and inhibiting myocardial apoptosis through the Akt-mediated signaling pathway.


Subject(s)
Genetic Therapy/methods , Heart/drug effects , Kallikreins/administration & dosage , Myocardial Infarction/therapy , Protein Serine-Threonine Kinases , Ventricular Remodeling/drug effects , Adenoviridae/genetics , Animals , Apoptosis/drug effects , Cell Survival , Disease Models, Animal , Dobutamine , Gene Transfer Techniques , Heart/physiopathology , Heart Function Tests/drug effects , Hemodynamics/drug effects , Humans , Kallikrein-Kinin System/physiology , Kallikreins/genetics , Ligation , Male , Myocardial Infarction/diagnosis , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocardium/pathology , Nitrates/metabolism , Nitrites/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Wistar , Treatment Outcome
12.
Am J Physiol Endocrinol Metab ; 283(6): E1291-8, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12424108

ABSTRACT

Adrenomedullin (AM) is a potent vasodilating peptide and is involved in cardiovascular and renal disease. In the present study, we investigated the role of AM in cardiac and renal function in streptozotocin (STZ)-induced diabetic rats. A single tail-vein injection of adenoviral vectors harboring the human AM gene (Ad.CMV-AM) was administered to the rats 1-wk post-STZ treatment (65 mg/kg iv). Immunoreactive human AM was detected in the plasma and urine of STZ-diabetic rats treated with Ad.CMV-AM. Morphological and chemical examination showed that AM gene delivery significantly reduced glycogen accumulation within the hearts of STZ-diabetic rats. AM gene delivery improved cardiac function compared with STZ-diabetic rats injected with control virus, as observed by decreased left ventricular end-diastolic pressure, increased cardiac output, cardiac index, and heart rate. AM gene transfer significantly increased left ventricular long axis (11.69 +/- 0.46 vs. 10.31 +/- 0.70 mm, n = 10, P < 0.05) and rate of pressure rise and fall (+6,090.1 +/- 597.3 vs. +4,648.5 +/- 807.1 mmHg/s), (-4,902.6 +/- 644.2 vs. -3,915.5 +/- 805.8 mmHg/s, n = 11, P < 0.05). AM also significantly attenuated renal glycogen accumulation and tubular damage in STZ-diabetic rats as well as increased urinary cAMP and cGMP levels, along with increased cardiac cAMP and Akt phosphorylation. We also observed that delivery of the AM gene caused an increase in body weight along with phospho-Akt and membrane-bound GLUT4 levels in skeletal muscle. These results suggest that AM plays a protective role in hyperglycemia-induced glycogen accumulation and cardiac and renal dysfunction via Akt signal transduction pathways.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/prevention & control , Heart/drug effects , Muscle Proteins , Peptides/pharmacology , Protein Serine-Threonine Kinases , Adenoviridae/genetics , Adrenomedullin , Animals , Blood Glucose , Body Weight/drug effects , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/complications , Disease Models, Animal , Gene Expression , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Glucose Transporter Type 4 , Glycogen/metabolism , Heart/physiopathology , Heart Function Tests/drug effects , Humans , Kidney/cytology , Kidney/drug effects , Kidney/pathology , Male , Monosaccharide Transport Proteins/metabolism , Muscle, Skeletal/metabolism , Myocardium/metabolism , Myocardium/pathology , Peptides/genetics , Peptides/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , Streptozocin , Ventricular Function, Left/drug effects
13.
Blood ; 100(9): 3245-52, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12384424

ABSTRACT

Kallistatin is a unique serine proteinase inhibitor (serpin) and a heparin-binding protein. It has been localized in vascular smooth muscle cells and endothelial cells of human blood vessels, suggesting that kallistatin may be involved in the regulation of vascular function. Our previous study showed that kallistatin plays a role in neointima hyperplasia. In this study, we investigated the potential role of kallistatin in angiogenesis in vitro and in vivo. Purified human kallistatin significantly inhibited vascular endothelial growth factor (VEGF)- or basic fibroblast growth factor (bFGF)-induced proliferation, migration, and adhesion of cultured endothelial cells. Kallistatin attenuated VEGF- or bFGF-induced capillary density and hemoglobin content in subcutaneously implanted Matrigel plugs in mice. To further investigate the role of kallistatin in angiogenesis, we prepared adenovirus carrying the human kallistatin cDNA (Ad.HKBP) and evaluated the effect of kallistatin gene delivery on spontaneous angiogenesis in a rat model of hind-limb ischemia. Local kallistatin gene delivery significantly reduced capillary formation and regional blood perfusion recovery in the ischemic hind limb after removal of the femoral artery. Furthermore, a single intratumoral injection of Ad.HKBP into pre-established human breast tumor xenografts grown in athymic mice resulted in significant inhibition of tumor growth. CD31 immunostaining of tumor sections showed a decreased number of blood vessels in the kallistatin-treated group as compared to the control. These results demonstrate a novel role of kallistatin in the inhibition of angiogenesis and tumor growth.


Subject(s)
Angiogenesis Inhibitors/physiology , Carrier Proteins/physiology , Serpins/physiology , Adenocarcinoma/pathology , Adenoviridae/genetics , Angiogenesis Inhibitors/genetics , Animals , Breast Neoplasms/pathology , Carrier Proteins/genetics , Cell Adhesion , Cell Division , Cell Movement , Cells, Cultured , DNA, Complementary/administration & dosage , DNA, Complementary/genetics , Drug Implants , Endothelial Growth Factors/administration & dosage , Endothelial Growth Factors/antagonists & inhibitors , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Female , Fibroblast Growth Factor 2/administration & dosage , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/pharmacology , Genetic Vectors/administration & dosage , Hindlimb/blood supply , Humans , Injections, Intralesional , Intercellular Signaling Peptides and Proteins/administration & dosage , Intercellular Signaling Peptides and Proteins/pharmacology , Ischemia/physiopathology , Lymphokines/administration & dosage , Lymphokines/antagonists & inhibitors , Lymphokines/pharmacology , Male , Neovascularization, Pathologic/therapy , Neovascularization, Physiologic/drug effects , Rats , Recombinant Fusion Proteins/physiology , Serpins/genetics , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Xenograft Model Antitumor Assays
14.
Arterioscler Thromb Vasc Biol ; 22(8): 1279-85, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12171788

ABSTRACT

OBJECTIVE: Endothelium-derived NO has been shown to mediate the mitogenic effect of vascular endothelial growth factor on cultured microvascular endothelium. To evaluate the role of endothelial NO synthase (eNOS) in angiogenesis in the ischemic hindlimb, we engineered an adenovirus containing human eNOS cDNA. METHODS AND RESULTS: After gene transfer, expression of eNOS in cultured cells was detected by increased intracellular cGMP and nitrate/nitrite levels and NO synthase activity. Adenovirus containing either the eNOS or luciferase gene was injected into the adductor muscle of rat hindlimbs immediately after femoral artery removal. Human eNOS protein was detected throughout the course of the experiment by immunostaining. Significant increases in blood perfusion were monitored by laser Doppler imaging from 2 to 4 weeks after gene delivery in the ischemic hindlimb of rats receiving eNOS compared with control rats receiving the reporter gene. An increase in regional blood flow was also detected after eNOS gene transfer by a fluorescent microsphere assay. eNOS gene delivery in the ischemic hindlimb resulted in significant increases in intracellular cGMP levels and in capillary density identified by anti-CD-31 immunostaining. Angiogenesis was further confirmed in mice after eNOS gene transfer by increased hemoglobin content in Matrigel implants. CONCLUSIONS: Taken together, these results indicate that eNOS enhances angiogenesis and raises the potential of eNOS gene transfer for modulation of vascular insufficiency.


Subject(s)
Endothelium, Vascular/enzymology , Ischemia/physiopathology , Neovascularization, Physiologic , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/physiology , Adenoviridae , Animals , Capillaries/physiology , Cells, Cultured , Cyclic GMP/biosynthesis , DNA, Viral , Gene Transfer Techniques , Genetic Vectors , Hemoglobins/analysis , Hindlimb/blood supply , Ischemia/enzymology , Laser-Doppler Flowmetry , Male , Mice , Models, Animal , Plasmids , Rats , Regional Blood Flow
SELECTION OF CITATIONS
SEARCH DETAIL
...