Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Neuropathol Appl Neurobiol ; 37(7): 738-52, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21366664

ABSTRACT

AIM: The function of brain (neuronal) peroxisome proliferator-activated receptor(s) γ (PPARγ) in the delayed degeneration and loss of neurones in the substantia nigra (SN) was studied in rats after transient occlusion of the middle cerebral artery (MCAO). METHODS: The PPARγ agonist, pioglitazone, or vehicle was infused intracerebroventricularly over a 5-day period before, during and 5 days after MCAO (90 min). The neuronal degeneration in the SN pars reticularis (SNr) and pars compacta (SNc), the analysis of the number of tyrosine hydroxylase-immunoreactive (TH-IR) neurones and the expression of the PPARγ in these neurones were studied by immunohistochemistry and immunofluorescence staining. The effects of PPARγ activation on excitotoxic and oxidative neuronal damage induced by glutamate and 6-hydroxydopamine were investigated in primary cortical neurones expressing PPARγ. RESULTS: Pioglitazone reduced the total and striatal infarct size, neuronal degeneration in both parts of the ipsilateral SN, the loss of TH-IR neurones in the SNc and increased the number of PPARγ-positive TH-IR neurones. Pioglitazone protected primary cortical neurones against oxidative and excitotoxic damage, prevented the loss of neurites and supported the formation of synaptic networks in neurones exposed to glutamate or 6-hydroxydopamine by a PPARγ-dependent mechanism. CONCLUSIONS: Activation of cerebral PPARγ confers neuroprotection after ischaemic stroke by preventing both, neuronal damage within the peri-infarct zone and delayed degeneration of neurones and neuronal death in areas remote from the site of ischaemic injury. Pioglitazone and other PPARγ agonists may be useful therapeutic agents to prevent progression of brain damage after cerebral ischaemia.


Subject(s)
Infarction, Middle Cerebral Artery/drug therapy , Ischemic Attack, Transient/drug therapy , Neurons/drug effects , Neuroprotective Agents/therapeutic use , PPAR gamma/metabolism , Substantia Nigra/drug effects , Thiazolidinediones/therapeutic use , Animals , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Ischemic Attack, Transient/metabolism , Ischemic Attack, Transient/pathology , Male , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Pioglitazone , Rats , Rats, Wistar , Substantia Nigra/metabolism , Substantia Nigra/pathology , Thiazolidinediones/pharmacology
2.
Autoimmunity ; 42(3): 183-97, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19301199

ABSTRACT

The MerTK receptor tyrosine kinase is an important negative regulator of dendritic cell function and is required to prevent B cell autoimmunity in vivo. It is not currently known however, if any causal relationship exists between these two aspects of MerTK function. We sought to determine if dendritic cells (DC) from mice lacking MerTK (mertk(- / - ) mice) have characteristics that may aid in the development of B cell autoimmunity. Specifically, we found that mertk(- / - ) mice contain an elevated number of splenic DC, and this population contains an elevated proportion of cells secreting the critical B cell pro-survival factor, B cell activating factor (BAFF). Elevated numbers of BAFF-secreting cells were also detected among mertk(- / - ) bone marrow-derived dendritic cell (BMDC) populations. This was observed in both resting BMDC, and BMDC stimulated with lipopolysaccharide (LPS) or treated with exogenous apoptotic cells. We also found that DC in general have a pro-survival effect on resting B cells in co-culture. However, despite containing more BAFF-secreting cells, mertk(- / - ) BMDC were not superior to C57BL/6 or baff-deficient BMDC at promoting B cell survival. Furthermore, using decoy receptors, we show that DC may promote B cell survival and autoimmunity through a BAFF-and a proliferation-inducing ligand-independent mechanism.


Subject(s)
B-Cell Activating Factor/metabolism , Dendritic Cells/metabolism , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Age Factors , Animals , Antibodies, Antinuclear/blood , Antibodies, Antinuclear/immunology , Autoimmunity/physiology , B-Cell Activating Factor/blood , B-Cell Activating Factor/pharmacology , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Count , Cell Differentiation/drug effects , Cell Survival/physiology , Chromatin/immunology , Coculture Techniques , DNA/immunology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells, Follicular/cytology , Dendritic Cells, Follicular/metabolism , Gene Expression/genetics , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Recombinant Proteins/pharmacology , Spleen/cytology , Spleen/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/physiology , c-Mer Tyrosine Kinase
3.
Ann Hum Genet ; 71(Pt 1): 1-7, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17227472

ABSTRACT

The strong association between the angiotensin I-converting enzyme (ACE) gene I/D polymorphism with serum ACE activity appears lacking in Nigerians and Kenyans, but has not previously been well assessed in others of African origin. This study addressed this issue in an ethnically well defined black South African population. A putative association for the A22982G ACE gene variant, a QTL likely to impact on serum ACE activity, was also sought. Subjects were 200 healthy male black South African volunteers from the Xhosa ethnic group. Venous blood was obtained from all subjects for DNA extraction. ACE I/D and A22982G genotypes were determined and serum ACE activity measured. Age and blood pressure were recorded. For the group as a whole (mean +/- SD age 38.5 +/- 9.8 years, SBP 119.6 +/- 14.1 mmHg, DBP 78.2 +/- 10.1 mmHg) serum ACE activity was 38.2 +/- 11.2 nmol ml(-1)min(-1). ACE I/D genotype was not significantly associated with serum ACE activity. In contrast, the A22982G variant was significantly associated with serum ACE activity, being 35.9 +/- 9.6, 38.1 +/- 10.6 and 42.4 +/- 15.3 nmol ml(-1)min(-1) for AA, AG and GG genotypes respectively; p = 0.03 by ANOVA and p = 0.01 by linear trend. In keeping with the findings in some other African populations, the ACE I/D polymorphism is not strongly associated with serum ACE activity in Xhosa South Africans. As such, it cannot be used as a marker of ACE activity in these subjects. In this regard the use of the A22982G gene variant may be more appropriate.


Subject(s)
Black People/genetics , Peptidyl-Dipeptidase A/blood , Peptidyl-Dipeptidase A/genetics , Blood Pressure , Genotype , Humans , Male , Polymorphism, Genetic , South Africa
4.
Eur J Appl Physiol ; 92(4-5): 579-83, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15048577

ABSTRACT

Angiotensin-converting enzyme (ACE) activity has been suggested as a determinant of some exercise phenotypes via some studies that have associated the ACE gene with exercise performance, although several studies provide conflicting evidence regarding the influence of the ACE gene. The relationships between ACE phenotype (ACE activity) and various exercise parameters should also be examined. An early step in this process is to determine whether common environmental stimuli such as exercise and diet have acute effects on ACE activity. In this study, the acute effects of aerobic exercise, resistance exercise and glucose ingestion on circulating ACE activity were examined. On three separate occasions, 20 healthy adult volunteers (9 female and 11 male) performed 20 min of submaximal cycle exercise at 70-80% of maximal heart rate, four sets of ten repetitions of unilateral leg extension resistance exercise at ten-repetition maximum load, or ingested 1 g kg(-1) glucose. Circulating ACE activity was assessed for 1 h after each intervention using a modified fluorometric method. Pre-intervention ACE activity remained remarkably stable across test days (difference < or =1.8%). Furthermore, there was no significant change in circulating ACE activity following any of the interventions (difference from pre-intervention values < or =6.8% when unadjusted for plasma volume changes, < or =4.5% when adjusted for plasma volume changes). These results suggest that acute exercise and glucose ingestion interventions as used here do not affect circulating ACE activity. These findings are an early step in illuminating the relationships between ACE activity and various exercise parameters.


Subject(s)
Exercise/physiology , Glucose/pharmacology , Peptidyl-Dipeptidase A/blood , Adult , Female , Genotype , Humans , Male , Peptidyl-Dipeptidase A/genetics , Physical Endurance/physiology , Plasma Volume/physiology , Weight Lifting/physiology
5.
J Hum Hypertens ; 16 Suppl 3: S64-70, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12140731

ABSTRACT

Biochemical, physiological and functional studies suggest that the brain renin-angiotensin system (RAS) is regulated independently of the peripheral RAS. The classical actions of angiotensin II in the brain include blood pressure control, drinking behaviour, natriuresis and the release of vasopressin into the circulation. At least two subtypes of G-protein coupled receptors, the AT(1) and the AT(2) receptor, have been identified. Most of the classic actions of angiotensin II in the brain are mediated by AT(1) receptors. The AT(2) receptor is involved in brain development and neuronal regeneration and protection. Additionally, AT(2) receptors can modulate some of the classic angiotensin II actions in the brain. Selective non-peptide AT(1) receptor blockers, applied systemically, have been shown to inhibit both peripheral and brain AT(1)receptors. In genetically hypertensive rats, inhibition of brain AT(1) receptors may contribute to the blood pressure lowering effects of AT(1) receptor blockers. Animal studies have shown that AT(1) receptor antagonists enable endogenous angiotensin II to stimulate neuronal regeneration via activation of AT(2) receptors. In animal models, inhibition of the brain RAS proved to be beneficial with respect to stroke incidence and outcome. Blockade of brain and cerebrovascular AT(1) receptors by AT(1) receptor blockers prevents the reduction in blood flow during brain ischaemia, reduces the volume of ischaemic injury and improves neurological outcome after brain ischaemia. This paper reviews the actions of angiotensin II and its receptors in the brain, and discusses the possible consequences of AT(1) receptor blockade in neuroprotection, neuroregeneration, cerebral haemodynamics and ischaemia.


Subject(s)
Angiotensin Receptor Antagonists , Brain/metabolism , Renin-Angiotensin System/physiology , Animals , Brain Ischemia/drug therapy , Brain Ischemia/physiopathology , Cerebrovascular Circulation/drug effects , Receptor, Angiotensin, Type 1
6.
Naunyn Schmiedebergs Arch Pharmacol ; 365(6): 477-83, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12070762

ABSTRACT

In the brain, angiotensin II (Ang II) induces various effects such as blood pressure increase, the release of arginine vasopressin (AVP) and drinking behaviour. In the present study, we investigated the ability of the angiotensin II type-I (AT(1)) receptor antagonist, candesartan, administered peripherally, to block the central effects of Ang II. Experiments were performed in conscious rats instrumented with an intracerebroventricular (i.c.v.) cannula or a guide cannula into the paraventricular nucleus (PVN) and arterial and femoral catheters. Candesartan was administered intravenously (i.v.) at doses of 0.01, 0.1, 1 or 10 mg/kg. Controls received vehicle (0.05 N Na(2)CO(3)). The drinking response ( n=10-11 per group), the pressor response ( n=7-8) and the release of AVP into the circulation ( n=10-11) following i.c.v. Ang II (100 ng) were measured 0.5, 2, 4 and 24 h following i.v. drug application. Candesartan inhibited the central responses to i.c.v. injected Ang II dose- and time-dependently. At the highest dose (10 mg/kg), the drinking and pressor responses and the release of AVP in response to i.c.v. Ang II were completely blocked at 4 h and still markedly inhibited 24 h after the antagonist application (by 85%, 48% and 86%, respectively). The lowest dose of the antagonist was without effect. In a further experiment, the release of AVP induced by microinjection of Ang II (100 ng) into the PVN was determined before and 4 h after administration of vehicle or candesartan (1 mg/kg, i.v.). Candesartan completely blocked the AVP release into the circulation induced by Ang II microinjection into the PVN. Our results demonstrate that candesartan administered peripherally effectively inhibits responses mediated by AT(1) receptors localised in periventricular brain regions as well as inside the blood-brain-barrier.


Subject(s)
Angiotensin II/antagonists & inhibitors , Angiotensin II/pharmacology , Angiotensin Receptor Antagonists , Benzimidazoles/administration & dosage , Tetrazoles/administration & dosage , Animals , Arginine Vasopressin/antagonists & inhibitors , Arginine Vasopressin/blood , Biphenyl Compounds , Blood Pressure/drug effects , Blood Pressure/physiology , Dose-Response Relationship, Drug , Drinking/drug effects , Drinking/physiology , Male , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1 , Receptors, Angiotensin/physiology , Wakefulness/drug effects , Wakefulness/physiology
7.
J Pharmacol Exp Ther ; 298(1): 62-70, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11408526

ABSTRACT

The effects of systemic treatment with the AT1 receptor antagonist telmisartan on central effects of angiotensin II (Ang II), namely, increase in blood pressure, vasopressin release into the circulation, and drinking response, were investigated in conscious, normotensive rats. The central responses to i.c.v. Ang II (30 ng/kg) were measured at 0.5, 2, 4, and 24 h following acute i.v. or acute and chronic oral telmisartan application. At a dose of 10 mg/kg i.v., the drinking response to i.c.v. Ang II was completely blocked over 4 h, while the pressor response and the release of vasopressin in response to i.c.v. Ang II were blocked by 60 to 80%. The inhibition of the centrally mediated pressor and drinking response to Ang II was sustained over 24 h. The lower doses of telmisartan (0.3 and 1 mg/kg) significantly inhibited the Ang II-induced actions over 4 h. A consistent 24-h inhibition of the central responses to i.c.v. Ang II was obtained after acute and chronic oral treatment with 30 mg/kg telmisartan. Oral treatment with 1 and 3 mg/kg telmisartan produced a slight but inconsistent inhibition of the central actions of Ang II. Telmisartan concentrations measured in the cerebrospinal fluid following 8 days of consecutive daily oral treatment (1-30 mg/kg) ranged from 0.87 +/- 0.27 ng/ml (1 mg/kg/day) to 46.5 +/- 11.6 ng/ml (30 mg/kg/day). Our results demonstrate that, following peripheral administration, the AT1 receptor antagonist telmisartan can penetrate the blood-brain barrier in a dose- and time-dependent manner to inhibit centrally mediated effects of Ang II.


Subject(s)
Angiotensin II/antagonists & inhibitors , Angiotensin Receptor Antagonists , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Benzimidazoles/administration & dosage , Benzoates/administration & dosage , Blood-Brain Barrier/drug effects , Vasoconstrictor Agents/antagonists & inhibitors , Angiotensin II/pharmacology , Animals , Arginine Vasopressin/blood , Arginine Vasopressin/drug effects , Blood Pressure/drug effects , Blood Pressure/physiology , Blood-Brain Barrier/physiology , Drinking/drug effects , Drinking/physiology , Male , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/metabolism , Telmisartan , Vasoconstrictor Agents/pharmacology
8.
Clin Exp Hypertens ; 23(3): 177-87, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11339685

ABSTRACT

Short term treatments of normotensive Wistar Kyoto rats with angiotensin II (ANGII) or in combination with the AT1 receptor antagonist, losartan or PD123319, the AT2 receptor antagonist on systemic arterial blood pressure (MABP) and their influence on noradrenaline sensitivity in isolated mesenteric portal vein were evaluated. ANGII increased MABP as well as the contractile response to noradrenaline in vessels from ANGII-treated animals. MABP and the maximal effect of the concentration response curve for noradrenaline were prevented by losartan. However, PD123319 did not influence the blood pressure, but completely removed the vessels sensitivity to noradrenaline. ANGII combined with the AT1 and/or AT2 receptors blockade completely prevented the pressure response to ANGII, but the concentration response curve for noradrenaline did not differ from the vehicle-treated control curve. In conclusion both AT1- and AT2 receptor activation seems to be important in controlling noradrenaline sensitivity of rat portal vein smooth muscle.


Subject(s)
Norepinephrine/physiology , Portal Vein/physiology , Receptors, Angiotensin/physiology , Angiotensin Receptor Antagonists , Angiotensins/pharmacology , Animals , Antihypertensive Agents/pharmacology , Arteries/drug effects , Arteries/physiology , Blood Pressure/drug effects , Blood Pressure/physiology , Drug Resistance , Drug Synergism , Imidazoles/pharmacology , In Vitro Techniques , Losartan/pharmacology , Male , Norepinephrine/pharmacology , Pyridines/pharmacology , Rats , Rats, Inbred WKY , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Reference Values , Splanchnic Circulation/drug effects , Splanchnic Circulation/physiology , Vasoconstriction/drug effects , Vasoconstriction/physiology
9.
Cardiovasc Res ; 49(1): 110-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11121802

ABSTRACT

OBJECTIVE: Blockade of angiotensin AT(1) receptors has been shown to prevent cardiac remodeling and improve left ventricular function and survival after myocardial infarction (MI). However, the timing of initiation of treatment has not been fully elucidated. Therefore, the purpose of the present study was to compare the effects of very early (30 min after MI), early (3 and 24 h after MI) and delayed (7 days after MI) treatments with the angiotensin AT(1) receptor antagonist fonsartan (HR 720) on cardiac morphological and hemodynamic parameters in a rat model of MI-induced heart failure and to establish the therapeutic window for the start of treatment. METHODS: Male Wistar rats underwent coronary ligation and were randomized fonsartan (HR720) treatment starting 30 min, 3 h, 24 h and 7 days after MI or no treatment. Treatment was continued up to 6 weeks post MI. RESULTS: Fonsartan (HR720) treatment attenuated cardiac hypertrophy when treatment started 30 min or later after MI, limited infarct size when treatment initiated 3 and 24 h after MI, decreased left ventricular end-diastolic pressure when treatment started 3 h to 7 days after MI, and improved dP/dt(max) when treatment commenced 24 h and 7 days after MI compared to untreated infarct group. CONCLUSION: Our results show that angiotensin AT(1) receptor blockade with fonsartan (HR720) produced the best cardioprotective effects when treatment was started 3 to 24 h after MI although a start of treatment 7 days following MI still could improve functional parameters. These results suggest an optimal time window for the start of treatment with angiotensin AT(1) receptor antagonists seems to be between 3 and 24 h post MI.


Subject(s)
Angiotensin Receptor Antagonists , Biphenyl Compounds/administration & dosage , Heart Failure/prevention & control , Imidazoles/administration & dosage , Myocardial Infarction/complications , Animals , Biphenyl Compounds/therapeutic use , Drug Administration Schedule , Heart Failure/etiology , Heart Failure/pathology , Heart Failure/physiopathology , Hemodynamics/drug effects , Imidazoles/therapeutic use , Male , Myocardium/pathology , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1 , Survival Rate
10.
Cardiovasc Res ; 47(1): 57-67, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10869530

ABSTRACT

OBJECTIVES: Elevated systemic arterial blood pressure is associated with left ventricular hypertrophy and fibrosis. It has been suggested that both circulating and local myocardial renin-angiotensin systems play a role in mediating these responses. Here we describe the natural history of ventricular hypertrophy and fibrosis in the transgenic (mRen2)27 rat--a monogenetic model--which has a high tissue expression of the murine renin transgene, and suffers severe hypertension. We further explored the relative contribution of both hypertensive burden and circulating and tissue renin-angiotensin systems to the fibrotic process. METHODS: The transgenic rats were treated from 28 days old with (1) a hypotensive dose of the ACE inhibitor ramipril which inhibited both tissue and circulating ACE activity, (2) the calcium antagonist amlodipine, or (3) a non-hypotensive dose of ramipril which inhibited about 60% of tissue ACE activity with little effect on circulating ACE. Normotensive Sprague-Dawley rats were used as controls. RESULTS: The transgenics developed left ventricular hypertrophy along with perivascular and interstitial fibrosis which became progressively worse up to 24 weeks of age. Both the high dose of ramipril and amlodipine prevented the hypertrophy and fibrosis, whereas tissue ACE inhibition without lowering blood pressure had no effect, and actually led to a worsening of the fibrosis by 24 weeks. CONCLUSIONS: These results suggest that the development of left ventricular hypertrophy and fibrosis in the transgenic (mRen2)27 rat are regulated by blood pressure and not activity of the renin-angiotensin systems and that progression of fibrosis at 24 weeks involves a mechanism unrelated to local renin-angiotensin activity.


Subject(s)
Hypertension/complications , Myocardium/pathology , Renin-Angiotensin System , Amlodipine/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Animals, Genetically Modified , Antihypertensive Agents/therapeutic use , Collagen/analysis , Fibrosis , Hypertension/metabolism , Hypertension/pathology , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/pathology , Male , Mice , Myocardium/chemistry , Peptidyl-Dipeptidase A/blood , Ramipril/therapeutic use , Rats , Rats, Sprague-Dawley , Time Factors
11.
Pharmacol Res ; 41(6): 641-8, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10816333

ABSTRACT

The aim of the study was to examine the influence of a short-term treatment of conscious Wistar Kyoto rats (WKY) and stroke-prone spontaneously hypertensive rats (SHRSP) by angiotensin II (ANG II) and by ANG II in combination with either l -NAME, HOE 140 or minoxidil on the mean arterial blood pressure (MABP) and the noradrenaline sensitivity in isolated portal vein preparations. MABP was significantly increased by ANG II treatment and ANG II plus l -NAME. However, it was slightly affected by ANG II in association with HOE 140, and significantly lowered by ANG II plus minoxidil. In control animals noradrenaline increased the frequency and the tone of contractile force. While ANG II enhanced the contractile response to noradrenaline, neither in combination with l -NAME, HOE 140 nor minoxidil prevented such an increase in the response to noradrenaline. In the presence of ergotamine, the contractile response to noradrenaline was completely blocked not only in control animals, but also in animals treated with ANG II alone or in combination with minoxidil. However, ergotamine (3 microm) failed to block completely the contractile response to noradrenaline in vessels from animals treated by ANG II in combination with l -NAME or HOE 140. These data suggest that ANG II causes an increase of noradrenaline sensitivity in the isolated portal vein of rat. l -NAME and HOE 140 unmask a contractile response to noradrenaline in the presence of ergotamine which seems to be mediated not only by alpha-adrenoceptors, but may be compensated by an endothelial relaxation.


Subject(s)
Angiotensin II/pharmacology , Hypertension/physiopathology , Nitric Oxide/antagonists & inhibitors , Norepinephrine/pharmacology , Vasoconstriction/drug effects , Angiotensin II/administration & dosage , Animals , Blood Pressure/drug effects , Bradykinin/administration & dosage , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Drug Interactions , Ergotamine/administration & dosage , Ergotamine/pharmacology , Hypertension/drug therapy , In Vitro Techniques , Male , Minoxidil/administration & dosage , Minoxidil/pharmacology , NG-Nitroarginine Methyl Ester/administration & dosage , NG-Nitroarginine Methyl Ester/pharmacology , Norepinephrine/administration & dosage , Portal Vein/drug effects , Portal Vein/physiopathology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Vasoconstriction/physiology
12.
Eur J Pharmacol ; 385(2-3): 171-9, 1999 Dec 03.
Article in English | MEDLINE | ID: mdl-10607873

ABSTRACT

Cardiac remodeling after myocardial infarction is associated with impaired ventricular function and heart failure and has important implications for survival. The purpose of the present study was to assess the effects of chronic treatment with a novel angiotensin AT(1) receptor antagonist 2-butyl-4-(methylthio-)-1-[[2'[[[(propylamino)carbonyl]amino]sulfonyl ](1,1'-biphenyl)-4-yl]methyl]-1H-imidazole-5-carboxylate (HR720), on cardiac remodeling and left ventricular dysfunction in a rat model of large myocardial infarction. Rats were subjected to permanent ligation of the left coronary artery and were treated for six weeks with placebo or HR720 (3 mg/kg/day) initiated 24 h after surgery. Sham-operated rats served as normal controls. Mean arterial blood pressure, the maximum rate of rise of the left ventricular systolic pressure (dP/dt(max)), left ventricular end-diastolic pressure, left ventricular inner diameter and circumference, septal thickness, left ventricular collagen content and heart weight were measured at the end of the treatment. HR720 treatment versus placebo attenuated the cardiac hypertrophy (heart weight/body weight: 2.88+/-0.08 mg/g vs. 3.16+/-0.09 mg/g, P<0.05), reduced interstitial collagen content (3. 47+/-0.28% vs. 5.25+/-0.45%, P<0.01), limited infarct size (33.0+/-3. 0% vs. 41.5+/-2.3%, P<0.05), decreased left ventricular end-diastolic pressure (13.7+/-2.2 vs. 21.4+/-1.6 mm Hg, P<0.01) and improved dP/dt(max) (9000+/-430 vs. 6000+/-840 mm Hg/s, P<0.05). The present results demonstrate that chronic treatment with the angiotensin AT(1) receptor antagonist HR720 can limit infarct size, partially prevent cardiac hypertrophic remodeling and improve left ventricular function in rats with myocardial infarction.


Subject(s)
Angiotensin Receptor Antagonists , Biphenyl Compounds/pharmacology , Imidazoles/pharmacology , Myocardial Infarction/prevention & control , Animals , Blood Pressure/drug effects , Body Weight/drug effects , Cardiomegaly/prevention & control , Heart Rate/drug effects , Heart Ventricles/drug effects , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Hemodynamics/drug effects , Male , Myocardial Contraction/drug effects , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Ventricular Function, Left/drug effects
13.
Am J Physiol ; 277(6): H2333-40, 1999 12.
Article in English | MEDLINE | ID: mdl-10600853

ABSTRACT

A reduction of renal kallikrein has been found in non-insulin-treated diabetic individuals, suggesting that an impaired renal kallikrein-kinin system (KKS) contributes to the development of diabetic nephropathy. We analyzed relevant components of the renal KKS in non-insulin-treated streptozotocin (STZ)-induced diabetic rats. Twelve weeks after a single injection of STZ, rats were normotensive and displayed hyperglycemia, polyuria, proteinuria, and reduced glomerular filtration rate. Blood bradykinin (BK) levels and prekallikrein activity were significantly increased compared with controls. Renal kallikrein activity was reduced by 70%, whereas urinary BK levels were increased up to threefold. Renal kininases were decreased as indicated by a 3-fold reduction in renal angiotensin-converting enzyme activity and a 1.8-fold reduction in renal expression of neutral endopeptidase 24.11. Renal cortical expression of kininogen and B2 receptors was enhanced to 1.4 and 1. 8-fold, respectively. Our data suggest that increased urinary BK levels found in severely hyperglycemic STZ-diabetic rats are related to increased filtration of components of the plasma KKS and/or renal kininogen synthesis in combination with decreased renal kinin-degrading activity. Thus, despite reduced renal kallikrein synthesis, renal KKS is activated in the advanced stage of diabetic nephropathy.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/physiopathology , Kallikreins/metabolism , Kidney/physiopathology , Kinins/metabolism , Animals , Bradykinin/blood , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Glomerular Filtration Rate/drug effects , Insulin/pharmacology , Kidney/metabolism , Kidney Cortex/metabolism , Kidney Medulla/metabolism , Male , Peptidyl-Dipeptidase A/metabolism , Polyuria , Prekallikrein/metabolism , Proteinuria , Rats , Rats, Wistar , Reference Values
14.
Drugs Today (Barc) ; 35(2): 105-15, 1999 Feb.
Article in English | MEDLINE | ID: mdl-12973413

ABSTRACT

The nonpeptide AT(1) receptor antagonist candesartan is generated from the prodrug candesartan cilexetil during gastrointestinal absorption. In vitro studies have shown that candesartan acts as an insurmountable, tightly bound antagonist at the AT(1) receptor, producing a dose-dependent reduction in the maximal responses to angiotensin II (AII) and virtually an elimination of the AT(1) receptor-mediated effects of AII at high concentrations. The binding of candesartan to the AT(1) receptor is highly selective, and the drug dissociates slowly from the receptor. Candesartan cilexetil produces the expected changes in the parameters of the renin-angiotensin system. Plasma renin activity and plasma AII concentrations were increased and aldosterone levels decreased following drug application. As a consequence, stimulation of AT(2) receptor-mediated actions of AII, such as growth inhibition and vasodilation, may contribute to the overall effects of the AT(1) antagonist, since the AT(2) receptors are left unopposed by candesartan. The antihypertensive efficacy of candesartan cilexetil has been demonstrated in different animal models of hypertension including 2 kidney-1 clip and 1 kidney-1 clip hypertensive rats and spontaneously hypertensive rats (SHR). Candesartan cilexetil produced a slow onset, long-lasting antihypertensive action at a dose range of 0.1-10 mg/kg with no rebound effect upon drug withdrawal. A growing number of studies indicate that candesartan cilexetil can produce end organ protection in addition to lowering blood pressure. In preclinical studies, candesartan cilexetil caused prevention and regression of left ventricular hypertrophy and cardiac fibrosis, protected the heart against ischemia-reperfusion injury and reduced myocardial damage during myocarditis. In different animal models of renal dysfunction, candesartan cilexetil reduced proteinuria and albuminuria, inhibited histopathological renal changes and controlled the renal expression of TGF-beta1 and collagen types I and III. Finally, in stroke prone SHR, candesartan cilexetil markedly attenuated the incidence of stroke even at low doses, with minimal blood pressure lowering effects, and fully protected against stroke at higher doses.

15.
J Hypertens Suppl ; 16(7): S3-9, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9855025

ABSTRACT

BACKGROUND: The renin-angiotensin system is a widely studied hormonal system that comprises substrate-enzyme interactions, the end result of which is the production of the active peptide angiotensin II. Because angiotensin II affects blood pressure control, sodium and water homeostasis, and cardiovascular function and structure, a great deal of research effort has been directed toward blocking the renin-angiotensin system. Angiotensin II also may be involved in end-organ damage in hypertension, heart failure, and vascular disease. ANGIOTENSIN II RECEPTORS: At least two subtypes of angiotensin II receptors have been identified, angiotensin type 1 (AT)1 and type 2 (AT2). The AT1 receptor mediates all the known actions of angiotensin II on blood pressure control. Additionally, research has indicated that the AT1 receptor modulates cardiac contractility and glomerular filtration, increases renal tubular sodium reabsorption, and cardiac and vascular hypertrophy. Less is known about the function of the AT2 receptor. Evidence suggests that the AT2 receptor inhibits cell proliferation and reverses the AT1-induced hypertrophy. Indeed, these receptors are thought to exert opposing effects. ANGIOTENSIN RECEPTOR ANTAGONISTS: This newly introduced class of drugs is able to inhibit the renin-angiotensin system at the receptor level by specifically blocking the AT1 receptor subtype. These drugs induce a dose-dependent blockade of angiotensin II effects, resulting in reduced blood pressure, urinary protein, and glomerular sclerosis. It is postulated that AT1 receptor antagonists may provide end-organ protection by blocking angiotensin II effects via the AT1 receptor, leaving the AT2 receptor unopposed. Consequently, these agents may reduce the morbidity and mortality that result from myocardial infarction and other conditions resulting from structural alterations in the heart, kidney, and vasculature.


Subject(s)
Angiotensin Receptor Antagonists , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Cardiovascular Diseases/drug therapy , Renin-Angiotensin System/drug effects , Blood Pressure/drug effects , Blood Vessels/innervation , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Heart/innervation , Humans , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Myocardial Contraction/drug effects , Ventricular Function, Left/drug effects
16.
J Hypertens ; 16(5): 635-43, 1998 May.
Article in English | MEDLINE | ID: mdl-9797175

ABSTRACT

BACKGROUND: Activation of the renin-angiotensin system has been implicated strongly in the transition from benign to malignant hypertension. However, the concomitant rise in blood pressure might also have a direct effect on the vascular wall by initiating fibrinoid necrosis and myointimal proliferation. Ascertaining the relative importance of these two factors in this process has proved difficult. TGR(mREN2)27 heterozygotes (HanRen2/Edin- -) have previously been shown to develop malignant hypertension spontaneously and exhibit the characteristic features of human malignant hypertension. OBJECTIVE: Tissue renin-angtiotensin systems have been implicated in the pathogenesis of malignant hypertension. We set out to determine whether inhibition of this system might protect against development of the disease in a rat model. METHOD: Male TGR(mREN2)27 heterozygotes (n = 24) were given a non-hypotensive dose of the angiotensin converting enzyme inhibitor ramipril (5 microg/kg per day) from 28 to 120 days of age, untreated rats acting as controls (n = 40). The incidences of malignant hypertension were compared. Systolic blood pressure was measured by tail-cuff plethysmography during treatment; tissue and plasma angiotensin converting enzyme levels and renal histological changes were assessed at the end of the treatment period or upon development of malignant hypertension. RESULTS: Sixty-three per cent of control rats and 4% of angiotensin converting enzyme inhibitor-treated rats had developed malignant hypertension by 120 days despite there having been no significant difference in systolic blood pressure throughout the course of treatment. Angiotensin converting enzyme activities in kidney, heart and resistance vessels, though not that in plasma, were significantly lower in the treated rats. The degree of medial wall thickening did not differ between the two groups whereas evidence of tissue injury (e.g. intimal fibrosis, fibrinoid necrosis and nephron injury) was significantly less common among rats in the angiotensin converting enzyme inhibitor-treated group. CONCLUSIONS: Tissue angiotensin converting enzyme inhibition at a non-hypotensive dose almost completely prevented mortality from malignant hypertension and significantly reduced tissue injury in this model, implicating angiotensin II rather than high blood pressure as the principal 'vasculotoxic' agent in malignant hypertension.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Hypertension/enzymology , Peptidyl-Dipeptidase A/metabolism , Ramipril/administration & dosage , Animals , Animals, Genetically Modified , Blood Pressure/drug effects , Heterozygote , Humans , Hypertension/genetics , Hypertension/prevention & control , Kidney/physiopathology , Male , Rats , Renin-Angiotensin System/physiology
17.
Biochem Biophys Res Commun ; 243(1): 142-7, 1998 Feb 04.
Article in English | MEDLINE | ID: mdl-9473495

ABSTRACT

A substantial increase in [125I]Sar1, Ile-Angiotensin II binding activity can be observed 24 hours after treatment of R3T3 cells with AT2 receptor agonists and antagonists. An increase in the radioligand binding activity, although less profound, can also be observed 6 hours after AT2 receptor ligand treatment, on fetal human kidney cells expressing a recombinant human AT2 receptor. However, the increase in radioligand binding activity cannot be detected unless the ligands are removed from the cell surface by an acid-glycine (pH 3) wash, just prior to the binding assay. Interestingly, an acid-glycine wash 24 hours prior to the binding assay causes a dramatic decrease in the radioligand binding activity on untreated R3T3 cells. This decrease, which was prevented by angiotensin II treatment, suggests the existence of an unknown endogenous factor which, like the AT2 receptor ligands, seems to prevent AT2 receptor degradation.


Subject(s)
Receptors, Angiotensin/metabolism , 1-Sarcosine-8-Isoleucine Angiotensin II/metabolism , 3T3 Cells , Acids , Angiotensin II/metabolism , Angiotensin Receptor Antagonists , Animals , Cell Line , Glycine , Humans , Ligands , Mice , Radioligand Assay , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/agonists , Recombinant Proteins/metabolism
18.
Hypertension ; 31(1 Pt 2): 349-55, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9453327

ABSTRACT

In the present study we tested the hypothesis whether an angiotensin AT2 receptor-mediated stimulation of the bradykinin (BK)/nitric oxide (NO) system can account for the effects of AT1 receptor antagonism on aortic cGMP described previously in SHRSP. Adult SHRSP were treated for 4 hours with angiotensin II (ANG II) (30 ng/kg per min IV) or vehicle (0.9% NaCl I.V.). Animals were pretreated with vehicle, losartan (100 mg/kg P.O.), PD 123319 (30 mg/kg I.V.), losartan plus PD 123319, icatibant (500 microg/kg I.V.), N(G)-nitro-L-arginine methyl ester (L-NAME; 1 mg/kg I.V.), or minoxidil (3 mg/kg I.V.). Mean arterial blood pressure (MAP) was continuously monitored over the 4-hour experimental period, and plasma ANG II and aortic cGMP were measured by RIA at the end of the study. ANG II infusion over 4 hours raised MAP by about 20 mm Hg. Losartan alone or losartan plus ANG II as well as minoxidil plus ANG II markedly reduced blood pressure when compared to vehicle-treated or ANG II-treated animals, respectively. Plasma levels of ANG II were increased 2-fold by ANG II infusion alone or by ANG II in combination with icatibant, L-NAME, or minoxidil. The increase in plasma ANG II levels was even more pronounced after losartan treatment. Aortic cGMP content was significantly increased by ANG II, losartan, losartan plus ANG II, and minoxidil plus ANG II by 60%, 45%, 68%, and 52%, respectively (P<.05). The effects of ANG II and of losartan plus ANG II on aortic cGMP content were both blocked by cotreatment with the AT2 receptor antagonist PD 123319. Icatibant and L-NAME abolished the effects of ANG II on aortic cGMP. Our results demonstrate the following: (1) ANG II increases aortic cGMP by an AT2 receptor-mediated action because the effect could be prevented by an AT2 receptor antagonist; (2) the effect of ANG II was not secondary to blood pressure increase because it remained under reduction of MAP with minoxidil; (3) losartan increased aortic cGMP most likely by increasing plasma ANG II levels with a subsequent stimulation of AT2 receptors; and (4) the effects of AT2 receptor stimulation are mediated by BK and, subsequently, NO because they were abolished by B2 receptor blockade as well as by NO synthase inhibition.


Subject(s)
Angiotensin II/pharmacology , Aorta/metabolism , Blood Pressure/drug effects , Cyclic GMP/metabolism , Hypertension/physiopathology , Losartan/pharmacology , Muscle, Smooth, Vascular/metabolism , Receptors, Angiotensin/physiology , Angiotensin II/blood , Animals , Aorta/drug effects , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Bradykinin Receptor Antagonists , Cerebrovascular Disorders , Hypertension/metabolism , Imidazoles/pharmacology , Male , Minoxidil/pharmacology , Muscle, Smooth, Vascular/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Pyridines/pharmacology , Rats , Rats, Inbred SHR , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/drug effects , Reference Values
19.
Am J Cardiol ; 80(3A): 110A-117A, 1997 Aug 04.
Article in English | MEDLINE | ID: mdl-9293963

ABSTRACT

Left ventricular hypertrophy (LVH) is considered to be an independent risk factor giving rise to ischemia, arrhythmia, and left ventricular dysfunction. In this article, we summarize recent studies performed in our laboratory to investigate (1) the contribution of the renin-angiotensin system to the cardiac remodeling process, which is triggered by myocardial infarction (MI) or hypertension-induced cardiac hypertrophy; (2) the effects of angiotensin-converting enzyme (ACE) inhibition and angiotensin AT1 receptor antagonism on cardiac parameters, such as myocardial infarct size, cardiac hypertrophy, heart function, and myocardial metabolism; (3) the mechanism of an ACE inhibitor-induced increase in cardiac capillary density in spontaneously hypertensive rats (SHR) and stroke prone SHR (SHR-SP). We observed that AT1 receptor gene expression in rat vascular smooth muscle cells (but not in rat coronary endothelial cells) was markedly enhanced after an ischemic insult in vitro. In a rat model in which MI was induced by coronary artery ligation, the AT1 receptor mRNA levels were transiently increased after MI and reached a peak level 24 hours post-MI. The AT2 receptor gene expression increased in a pattern similar to that of the AT1 receptor. ACE expression at the protein level in the repairing scar, which was demonstrated by monoclonal antibody staining, started to increase 2 weeks after MI and reached a peak level 3 weeks post-MI. Furthermore, long-term treatment with an ACE inhibitor limited infarct size, prevented cardiac hypertrophy, and improved heart function in the rat MI model. In SHR-SP, long-term treatment with either an ACE inhibitor or an AT1 receptor antagonist improved cardiac function and metabolism. Cardiac metabolism was even improved after low-dose ACE inhibitor treatment, which did not prevent hypertension and cardiac hypertrophy. In both SHR and SHR-SP, we found that the ACE inhibitor ramipril significantly increased capillary length density independently of its antihypertensive and antihypertrophic actions. Most of the cardiac effects of the ACE inhibitor could be abolished by a bradykinin B2 receptor antagonist. Thus, these cardiac effects of ACE inhibitors can be ascribed, at least under our experimental conditions, to ACE inhibitor-induced bradykinin potentiation.


Subject(s)
Angiotensin Receptor Antagonists , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Hypertrophy, Left Ventricular/physiopathology , Myocardial Infarction/pathology , Myocardium/metabolism , Renin-Angiotensin System/drug effects , Animals , Fibrosis , Humans , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/pathology , Myocardial Infarction/physiopathology
20.
J Card Fail ; 3(2): 133-48, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9220313

ABSTRACT

Kinins are potent bioactive peptides formed by the enzymatic action of kallikrein on kininogens. The discovery that angiotensin-converting enzyme, which generates angiotensin II, is also a major degrading enzyme of kinins, gave rise to the hypothesis that kinin potentiation, in addition to angiotensin II reduction, may be involved in the therapeutic actions of angiotensin-converting enzyme inhibitors. Angiotensin-converting enzyme inhibitors have become important drugs in the treatment of hypertension, congestive heart failure, postmyocardial infarction, and diabetic nephropathy. Although angiotensin II reduction appears to be the predominant mechanism of the antihypertensive effect of chronic angiotensin-converting enzyme inhibitor treatment, the role of kinins in the antihypertensive effects of angiotensin-converting enzyme inhibitors seems to be renin dependent and cannot be generalized for all models of hypertension. On the other hand, at least under experimental conditions, various cardioprotective effects of angiotensin-converting enzyme inhibitors appear to be due to the potentiation of endogenous kinins, including improved cardiac function, structural changes following myocardial ischemia, and induction of capillary growth in hypertension-induced left ventricular hypertrophy.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Heart/physiology , Kallikrein-Kinin System/physiology , Kinins/physiology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Heart/drug effects , Heart Ventricles/drug effects , Heart Ventricles/pathology , Humans , Hypertension/drug therapy , Myocardial Infarction/physiopathology , Myocardial Ischemia/physiopathology , Phosphotransferases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...