Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 3051, 2023 05 26.
Article in English | MEDLINE | ID: mdl-37236964

ABSTRACT

The kidney plays a key role in the correction of systemic acid-base imbalances. Central for this regulation are the intercalated cells in the distal nephron, which secrete acid or base into the urine. How these cells sense acid-base disturbances is a long-standing question. Intercalated cells exclusively express the Na+-dependent Cl-/HCO3- exchanger AE4 (Slc4a9). Here we show that AE4-deficient mice exhibit a major dysregulation of acid-base balance. By combining molecular, imaging, biochemical and integrative approaches, we demonstrate that AE4-deficient mice are unable to sense and appropriately correct metabolic alkalosis and acidosis. Mechanistically, a lack of adaptive base secretion via the Cl-/HCO3- exchanger pendrin (Slc26a4) is the key cellular cause of this derailment. Our findings identify AE4 as an essential part of the renal sensing mechanism for changes in acid-base status.


Subject(s)
Kidney , Membrane Transport Proteins , Mice , Animals , Kidney/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Acid-Base Equilibrium/physiology , Nephrons/metabolism , Sulfate Transporters/metabolism , Bicarbonates/metabolism , Chloride-Bicarbonate Antiporters
2.
Pflugers Arch ; 465(12): 1763-73, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23842739

ABSTRACT

Cardiac atrophy as a consequence of mechanical unloading develops following exposure to microgravity or prolonged bed rest. It also plays a central role in the reverse remodelling induced by left ventricular unloading in patients with heart failure. Surprisingly, the intracellular Ca(2+) transients which are pivotal to electromechanical coupling and to cardiac plasticity were repeatedly found to remain unaffected in early cardiac atrophy. To elucidate the mechanisms underlying the preservation of the Ca(2+) transients, we investigated Ca(2+) cycling in cardiomyocytes from mechanically unloaded (heterotopic abdominal heart transplantation) and control (orthotopic) hearts in syngeneic Lewis rats. Following 2 weeks of unloading, sarcoplasmic reticulum (SR) Ca(2+) content was reduced by ~55 %. Atrophic cardiac myocytes also showed a much lower frequency of spontaneous diastolic Ca(2+) sparks and a diminished systolic Ca(2+) release, even though the expression of ryanodine receptors was increased by ~30 %. In contrast, current clamp recordings revealed prolonged action potentials in endocardial as well as epicardial myocytes which were associated with a two to fourfold higher sarcolemmal Ca(2+) influx under action potential clamp. In addition, Cav1.2 subunits which form the pore of L-type Ca(2+) channels (LTCC) were upregulated in atrophic myocardium. These data suggest that in early cardiac atrophy induced by mechanical unloading, an augmented sarcolemmal Ca(2+) influx through LTCC fully compensates for a reduced systolic SR Ca(2+) release to preserve the Ca(2+) transient. This interplay involves an electrophysiological remodelling as well as changes in the expression of cardiac ion channels.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium/metabolism , Myocardium/pathology , Action Potentials , Animals , Atrophy/physiopathology , Heart Transplantation , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Ryanodine Receptor Calcium Release Channel/biosynthesis , Sarcoplasmic Reticulum/metabolism , Transplantation, Heterotopic
3.
Br J Pharmacol ; 152(7): 1070-80, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17934515

ABSTRACT

BACKGROUND AND PURPOSE: Rho-kinase (ROCK) has been implicated in the pathophysiology of altered vasoregulation leading to hypertension. Here we describe the pharmacological characterization of a potent, highly selective and orally active ROCK inhibitor, the derivative of a class of azaindoles, azaindole 1 (6-chloro-N4-{3,5-difluoro-4-[(3-methyl-1H-pyrrolo[2,3-b]pyridin-4-yl)oxy]-phenyl}pyrimidine-2,4-diamine). EXPERIMENTAL APPROACH: Pharmacological characterization of azaindole 1 was performed with human recombinant ROCK in vitro. Vasodilator activity was determined using isolated vessels in vitro and different animal models in vivo. KEY RESULTS: This compound inhibited the ROCK-1 and ROCK-2 isoenzymes with IC50 s of 0.6 and 1.1 nM in an ATP-competitive manner. Although ATP-competitive, azaindole 1 was inactive against 89 kinases (IC50>10 microM) and showed only weak activity against an additional 21 different kinases (IC50=1-10 microM). Only the kinases TRK und FLT3 were inhibited by azaindole 1 in the sub-micromolar range, albeit with IC50 values of 252 and 303 nM, respectively. In vivo, azaindole 1 lowered blood pressure dose-dependently after i.v. administration in anaesthetized normotensive rats. In conscious normotensive and spontaneously hypertensive rats azaindole 1 induced a dose-dependent decrease in blood pressure after oral administration without inducing a significant reflex increase in heart rate. In anaesthetized dogs, azaindole 1 induced vasodilatation with a moderately elevated heart rate. CONCLUSIONS AND IMPLICATIONS: Azaindole 1 is representative of a new class of selective and potent ROCK inhibitors and is a valuable tool for the elucidation of the role of ROCK in the cardiovascular system.


Subject(s)
Cardiovascular System/drug effects , Diamines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , rho-Associated Kinases/antagonists & inhibitors , Administration, Oral , Animals , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Binding Sites/drug effects , Blood Pressure/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Computer Simulation , Death-Associated Protein Kinases , Dogs , Dose-Response Relationship, Drug , Female , Humans , Injections, Intravenous , Male , Mice , Models, Animal , Models, Molecular , Organ Culture Techniques , Phosphorylation , Polymerase Chain Reaction/methods , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Rabbits , Rats , Rats, Inbred SHR , Rats, Wistar , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Time Factors , Vasodilator Agents/administration & dosage , Vasodilator Agents/chemistry , Vasodilator Agents/pharmacology , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
5.
Kidney Int ; 60(4): 1309-23, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11576345

ABSTRACT

BACKGROUND: Uremia is characterized by inadequately increased sympathetic activity. Sympathetic overactivity is involved in the genesis of hypertension in uremia, but its potential role on progression has not been well investigated. To address this issue, the effect of subantihypertensive doses of an alpha blocker and a beta blocker, and their combination on renal morphology and on albuminuria were investigated in the model of the subtotally nephrectomized rat. METHODS: Male Sprague-Dawley rats were subjected to surgical ablation (SNX) or sham operation (sham). Three days after surgery groups were treated either with phenoxybenzamine (PBZ, 5 mg/kg body weight/day), metoprolol (MET, 150 mg/kg body weight/day) or their combination (PBZ 2.5 mg/kg body weight/day + MET, 50 mg/kg body weight/day). Renal morphology was evaluated after 12 weeks by quantitative histology, immunohistochemistry, and electron microscopy. Urine albumin excretion and kidney endothelin-1 (ET-1), platelet-derived growth factor (PDGF), and transforming growth factor-beta (TGF-beta) mRNA expression were assessed. RESULTS: Systolic blood pressure was significantly higher in all SNX groups compared with sham-operated controls with no difference in the SNX groups. The number of glomeruli per left kidney was reduced from 30,904 +/- 3212 to 17,480 +/- 2341 by SNX (-43.5%). Mean glomerular volume increased from 2.63 +/- 0.7 in untreated sham operated to 4.11 +/- 0.48 microm 3 x 10(6) in untreated SNX (56.3%). The glomerulosclerosis index did not change in SNX + PBZ rats, but was significantly lower in SNX + MET (0.56 +/- 0.14) and particularly SNX + PBZ + MET rats (0.49 +/- 0.11) than in untreated SNX (0.74 +/- 0.24). Glomerular capillary length density (LV) as a sensitive index of capillary obliteration was significantly lower in SNX and almost normalized in the three intervention groups. The same was true for the mean podocyte number per glomerulus. Glomerular ultrastructure in SNX was largely preserved by all treatments. The albumin excretion rate was significantly higher in untreated SNX than in sham; it was significantly lower in all treated SNX groups. CONCLUSION: The beneficial effect of non-hypotensive doses of alpha and beta blockers and their combination on renal morphology and albuminuria in the model of renal ablation argue for a blood pressure-independent role of sympathetic overactivity in the genesis of progression. In addition, the beneficial effect of adrenergic receptor blockade indicates that a substantial part is not mediated by sympathetic cotransmitters such as adenosine 5'-triphosphate (ATP) and neuropeptide Y (NPY).


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Glomerulosclerosis, Focal Segmental/pathology , Albuminuria/urine , Animals , Disease Progression , Drug Combinations , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/urine , Immunohistochemistry , In Situ Hybridization , Kidney/drug effects , Kidney/pathology , Male , Microscopy, Electron , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Sprague-Dawley
6.
Am J Physiol Renal Physiol ; 280(6): F1062-71, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11352846

ABSTRACT

The time course of the autoregulatory response of renal blood flow (RBF) to a step increase in renal arterial pressure (RAP) was studied in conscious dogs. After RAP was reduced to 50 mmHg for 60 s, renal vascular resistance (RVR) decreased by 50%. When RAP was suddenly increased again, RVR returned to baseline with a characteristic time course (control; n = 15): within the first 10 s, it rose rapidly to 70% of baseline (response 1), thus already comprising 40% of the total RVR response. Thereafter, it increased at a much slower rate until it started to rise rapidly again at 20-30 s after the pressure step (response 2). After passing an overshoot of 117% at 43 s, RVR returned to baseline values. Similar responses were observed after RAP reduction for 5 min or after complete occlusions for 60 s. When tubuloglomerular feedback (TGF) was inhibited by furosemide (40 mg i.v., n = 12), response 1 was enhanced, providing 60% of the total response, whereas response 2 was completely abolished. Instead, RVR slowly rose to reach the baseline at 60 s (response 3). The same pattern was observed when furosemide was given at a much higher dose (>600 mg i.v.; n = 6) or in combination with clamping of the plasma levels of nitric oxide (n = 6). In contrast to RVR, vascular resistance in the external iliac artery after a 60-s complete occlusion started to rise with a delay of 4 s and returned to baseline within 30 s. It is concluded that, in addition to the myogenic response and the TGF, a third regulatory mechanism significantly contributes to RBF autoregulation, independently of nitric oxide. The three mechanisms contribute about equally to resting RVR. The myogenic response is faster in the kidney than in the hindlimb.


Subject(s)
Homeostasis/physiology , Renal Circulation/physiology , Animals , Blood Pressure/physiology , Consciousness , Diuretics/pharmacology , Dogs , Enzyme Inhibitors/pharmacology , Feedback/physiology , Furosemide/pharmacology , Homeostasis/drug effects , Kidney Glomerulus/physiology , Kidney Tubules/physiology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/blood , Nitric Oxide Donors/pharmacology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Renal Circulation/drug effects , Vascular Resistance/physiology
7.
J Neurosci ; 21(9): 3073-84, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11312292

ABSTRACT

Neurotrophin-4 (NT-4) is perhaps the still most enigmatic member of the neurotrophin family. We show here that NT-4 is expressed in neurons of paravertebral and prevertebral sympathetic ganglia, i.e., the superior cervical (SCG), stellate (SG), and celiac (CG) ganglion. Mice deficient for NT-4 showed a significant reduction (20-30%) of preganglionic sympathetic neurons in the intermediolateral column (IML) of the thoracic spinal cord. In contrast, neuron numbers in the SCG, SG, and CG were unchanged. Numbers of axons in the thoracic sympathetic trunk (TST) connecting the SG with lower paravertebral ganglia were also reduced, whereas axon numbers in the cervical sympathetic trunk (CST) were unaltered. Axon losses in the TST were paralleled by losses of synaptic terminals on SG neurons visualized by electron microscopy. Furthermore, immunoreactivity for the synaptic vesicle antigen SV2 was clearly reduced in the SG and CG. Levels of catecholamines and tyrosine hydroxylase immunoreactivity were dramatically reduced in the SG and the CG but not in the SCG. Despite this severe phenotype in the sympathetic system, blood pressure levels were not reduced and displayed a pattern more typical of deficits in baroreceptor afferents. Numbers of IML neurons were unaltered at postnatal day 4, suggesting a postnatal requirement for their maintenance. In light of these and previous data, we hypothesize that NT-4 provided by postganglionic sympathetic neurons is required for establishing and/or maintaining synapses of IML neurons on postganglionic cells. Impairment of synaptic connectivity may consequently reduce impulse flow, causing a reduction in transmitter synthesis in postganglionic neurons.


Subject(s)
Autonomic Fibers, Preganglionic/metabolism , Autonomic Nervous System Diseases/genetics , Ganglia, Sympathetic/metabolism , Nerve Growth Factors/deficiency , Spinal Cord/metabolism , Animals , Autonomic Fibers, Preganglionic/pathology , Autonomic Nervous System Diseases/complications , Axons/pathology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Catecholamines/deficiency , Catecholamines/metabolism , Cell Count , Ganglia, Sympathetic/pathology , Hypertension/etiology , Lysosomes/pathology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Presynaptic Terminals/pathology , RNA, Messenger/metabolism , Spinal Cord/pathology , Stellate Ganglion/metabolism , Stellate Ganglion/pathology , Superior Cervical Ganglion/metabolism , Superior Cervical Ganglion/pathology , Tyrosine 3-Monooxygenase/deficiency , Tyrosine 3-Monooxygenase/metabolism
8.
Circ Res ; 88(5): 483-90, 2001 Mar 16.
Article in English | MEDLINE | ID: mdl-11249871

ABSTRACT

Expression of the voltage-gated K(+) channel Kv2.1, a possible molecular correlate for the cardiac delayed rectifier current (I(K)), has recently been shown to vary between individual ventricular myocytes. The functional consequences of this cell-to-cell heterogeneity in Kv2.1 expression are not known. Using multiplex single-cell reverse transcriptase-polymerase chain reaction (RT-PCR), we detected Kv2.1 mRNA in 47% of isolated midmyocardial myocytes from the rat left ventricular free wall that were positive for alpha-myosin heavy chain mRNA (n=74). Whole-cell patch-clamp recordings demonstrated marked differences in the magnitude of I(K) (200 to 1450 pA at V(Pip)=40 mV) between individual myocytes of the same origin. Furthermore, the tetraethylammonium (TEA)-sensitive outward current (I(TEA)), known to be partly encoded by Kv2.1 in mice, revealed a wide range of current magnitudes between single cells (150 to 1130 pA at V(Pip)=40 mV). Combined patch-clamp recordings and multiplex single-cell RT-PCR analysis of the same myocytes, however, showed no differences in I(K) or I(TEA) magnitude or inactivation kinetics between myocytes expressing Kv2.1 mRNA and those that did not express Kv2.1 mRNA. In contrast, in all midmyocardial myocytes expressing the transient outward potassium current (I(to1)), Kv4 mRNA, which has been shown to underlie I(to1), was detected (n=10). These results indicate that I(K) heterogeneity among individual left ventricular myocytes cannot be explained by the distribution pattern of Kv2.1 mRNA. Other mechanisms besides Kv2.1 mRNA expression appear to determine magnitude and kinetics of I(K) in rat ventricular myocytes.


Subject(s)
Heart Ventricles/drug effects , Membrane Potentials/physiology , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , RNA, Messenger/metabolism , Animals , Delayed Rectifier Potassium Channels , Female , Gene Expression , Heart Ventricles/cytology , Myosin Heavy Chains/genetics , Patch-Clamp Techniques , Potassium Channels/genetics , RNA, Messenger/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Shab Potassium Channels , Ventricular Function
9.
J Physiol ; 530(Pt 3): 443-55, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11158275

ABSTRACT

The effect of cardiac hypertrophy on electrocardiogram (ECG), action potential duration (APD) and repolarizing K+ currents was investigated in epicardial, midmyocardial and endocardial myocytes isolated from the rat left ventricular free wall. Cardiac hypertrophy was induced by stenosis of the ascending aorta (AS), which led to an increased pressure load (+85 +/- 10 u1v1vZ mm11Z Hg) of the left ventricle; sham-operated animals served as controls. In ECG recordings from AS rats, the QTc interval was prolonged and the main vectors of the QRS complex and the T-wave pointed in opposite directions, indicating an abnormal sequence of repolarization. APD and K+ currents were recorded using the whole-cell patch-clamp technique. In the AS group, APD90 (90 % repolarization) was significantly prolonged in epicardial and midmyocardial, but not endocardial myocytes. Corresponding to the increase in APD, the magnitude of the transient outward K+ current (Ito1) was significantly smaller (-30 %) in epicardial and midmyocardial, but not endocardial myocytes. Inactivation and steady-state inactivation of Ito1 were not affected by hypertrophy. Recovery from inactivation was slightly prolonged in endocardial myocytes from AS rats. No differences in delayed rectifier currents (IK) or inwardly rectifying K+ currents (IK1) were detected between myocytes of the three regions of sham-operated or AS animals. However, both currents were reduced by AS. The present data show that cardiac hypertrophy caused by pressure overload leads to an increase in APD and a decrease in Ito1 primarily in epicardial and midmyocardial myocytes, which implies a major role of alterations in Ito1 for the reduced gradient in APD. The effects of AS on IK1 and IK may slightly counteract the decrease in APD gradient. The observed changes in APD and underlying ionic currents could well explain the alterations in repolarization observed in the ECG induced by cardiac hypertrophy.


Subject(s)
Action Potentials/physiology , Aortic Valve Stenosis/physiopathology , Heart/physiopathology , Hemodynamics , Potassium Channels/physiology , Ventricular Function, Left/physiology , Animals , Blood Pressure , Cardiomegaly/physiopathology , Cells, Cultured , Electrocardiography , Endocardium/physiopathology , Female , Heart/physiology , Heart Rate , Heart Ventricles , Myocardium/cytology , Myocardium/pathology , Pericardium/physiopathology , Rats , Rats, Sprague-Dawley , Time Factors
10.
Circ Res ; 87(11): E53-60, 2000 Nov 24.
Article in English | MEDLINE | ID: mdl-11090555

ABSTRACT

Large-conductance potassium (BK) channels in vascular smooth muscle cells (VSMCs) sense both changes in membrane potential and in intracellular Ca(2+) concentration. BK channels may serve as negative feedback regulators of vascular tone by linking membrane depolarization and local increases in intracellular Ca(2+) concentration (Ca(2+) sparks) to repolarizing spontaneous transient outward K(+) currents (STOCs). BK channels are composed of channel-forming BKalpha and auxiliary BKbeta1 subunits, which confer to BK channels an increased sensitivity for changes in membrane potential and Ca(2+). To assess the in vivo functions of this ss subunit, mice with a disrupted BKbeta1 gene were generated. Cerebral artery VSMCs from BKbeta1 -/- mice generated Ca(2+) sparks of normal amplitude and frequency, but STOC frequencies were largely reduced at physiological membrane potentials. Our results indicate that BKbeta1 -/- mice have an abnormal Ca(2+) spark/STOC coupling that is shifted to more depolarized potentials. Thoracic aortic rings from BKbeta1 -/- mice responded to agonist and elevated KCl with a increased contractility. BKbeta1 -/- mice had higher systemic blood pressure than BKbeta1 +/+ mice but responded normally to alpha(1)-adrenergic vasoconstriction and nitric oxide-mediated vasodilation. We propose that the elevated blood pressure in BKbeta1 -/- mice serves to normalize Ca(2+) spark/STOC coupling for regulating myogenic tone. The full text of this article is available at http://www.circresaha.org.


Subject(s)
Calcium Signaling/genetics , Hypertension/genetics , Hypertension/metabolism , Potassium Channels, Calcium-Activated , Potassium Channels/genetics , Potassium/metabolism , Signal Transduction/genetics , Animals , Cells, Cultured , Cerebral Arteries/metabolism , Female , Gene Targeting , Homozygote , In Vitro Techniques , Large-Conductance Calcium-Activated Potassium Channels , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Potassium Channels/deficiency , Potassium Channels/metabolism , Potassium Chloride/pharmacology , Protein Subunits , RNA, Messenger/metabolism , Sequence Analysis, DNA , Vasoconstriction/drug effects , Vasoconstriction/genetics , Vasodilator Agents/pharmacology
11.
Am J Physiol Regul Integr Comp Physiol ; 279(6): R2214-21, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11080088

ABSTRACT

Autonomic cardiovascular control was characterized in conscious, chronically catheterized mice by spectral analysis of arterial pressure (AP) and heart rate (HR) during autonomic blockade or baroreflex modulation of autonomic tone. Both spectra were similar to those obtained in humans, but at approximately 10x higher frequencies. The 1/f relation of the AP spectrum changed to a more shallow slope below 0.1-0.2 Hz. Coherence between AP and HR reached 0.5 or higher below 0.3-0.4 Hz and also above 2.5 Hz. Muscarinic blockade (atropine) or beta-adrenergic blockade (atenolol) did not significantly affect the AP spectrum. Atropine reduced HR variability at all frequencies, but this effect waned above 1 Hz. beta-Adrenergic blockade (atenolol) slightly enhanced the HR variability only above 1 Hz. alpha-Adrenergic blockade (prazosin) reduced AP variability between 0.05 and 3 Hz, most prominently at 0. 15-0.7 Hz. A shift of the autonomic nervous tone by a hypertensive stimulus (phenylephrine) enhanced, whereas a hypotensive stimulus (nitroprusside) depressed AP variability at 1-3 Hz; other frequency ranges of the AP spectrum were not affected except for a reduction below 0.4 Hz after nitroprusside. Variability of HR was enhanced after phenylephrine at all frequencies and reduced after nitroprusside. As with atropine, the reduction with nitroprusside waned above 1 Hz. In conclusion, in mice HR variability is dominated by parasympathetic tone at all frequencies, during both blockade and physiological modulation of autonomic tone. There is a limitation for further reduction but not for augmentation of HR variability from the resting state above 1 Hz. The impact of HR on AP variability in mice is confined to frequencies higher than 1 Hz. Limits between frequency ranges are proposed as 0.15 Hz between VLF (very low frequency range) and LF (low frequency range) and 1.5 Hz between LF and HF (high frequency range).


Subject(s)
Autonomic Nervous System/physiology , Blood Pressure/physiology , Cardiovascular Physiological Phenomena , Heart Rate/physiology , Adrenergic beta-Antagonists/pharmacology , Animals , Atenolol/pharmacology , Atropine/pharmacology , Autonomic Nervous System/drug effects , Blood Pressure/drug effects , Cardiovascular System/drug effects , Cardiovascular System/innervation , Consciousness , Female , Heart Rate/drug effects , Mice , Mice, Inbred C57BL , Muscarinic Antagonists/pharmacology , Nitroprusside/pharmacology , Phenylephrine/pharmacology
12.
J Physiol ; 527 Pt 3: 611-22, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10990545

ABSTRACT

Large (up to +400 %) transient ( approximately 20 s) increases of blood flow were observed in the external iliac arteries of resting conscious dogs (n = 10) in the absence of major alerting or muscular activity. At the same time arterial pressure (AP) fell slightly while heart rate (HR) rose. The vasodilatations were resistant to atropine, ganglionic, beta-adrenergic and NO-synthase inhibition, but were suppressed by spinal or general anaesthesia. Vasodilatations of similar appearance were elicited by an alerting sound; these were abolished by atropine. The spontaneous vasodilatations occurred simultaneously and their magnitudes were well correlated between both legs, but were not correlated to the amount of concomitant activation of the surface electromyogram. The duration of this activation almost never outlasted 10 s. The reactive hyperaemia observed after a total occlusion of the artery even for 16 s was not large enough to explain the size of the spontaneous vasodilatations. Occlusion during peak flow of the vasodilatations did not affect the size of the reactive hyperaemia. Spectral analysis made separately for data segments with and without vasodilatation revealed that the vasodilatations substantially enhanced the variability of AP and HR at frequencies below approximately 0.1 Hz. In conclusion, large coordinated skeletal muscle vasodilatations were identified in resting conscious dogs, which are initiated neurally, but not by sympathetic-cholinergic or nitroxidergic fibres and which do not show any clear correlation to muscular contraction. The vasodilatations substantially affect the regulation of skeletal muscle blood flow and explain a significant portion of AP and HR variability.


Subject(s)
Blood Pressure/physiology , Muscle, Skeletal/blood supply , Vasodilation/physiology , Animals , Arousal/physiology , Dogs , Ganglionic Blockers/pharmacology , Heart Rate/physiology , Hexamethonium/pharmacology , Hindlimb/blood supply , Iliac Artery/physiology , Muscle, Skeletal/innervation , Regional Blood Flow/drug effects , Regional Blood Flow/physiology , Synaptic Transmission/drug effects , Vasoactive Intestinal Peptide/physiology
13.
Cardiovasc Res ; 45(4): 900-12, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10728416

ABSTRACT

OBJECTIVE: The bradycardic agent zatebradine (UL-FS 49) reduces heart rate without negative inotropic or proarrhythmic effects. The aim was to experimentally characterize the influence of zatebradine on arterial baroreflex sensitivity (BRS) and heart rate variability (HRV) which are generally considered as estimates of vagal activity and have prognostic value in patients after myocardial infarction (MI). METHODS: Conscious rats were studied 3 days after left coronary artery ligation or sham-operation (SH). BRS was determined by linear regression analysis of RR-interval and mean arterial pressure changes evoked by intravenous (i.v.) injections of methoxamine and nitroprusside. HRV at rest was calculated from high-resolution electrocardiogram-recordings. RESULTS: In MI-rats heart rate was similar to SH-rats, mean arterial pressure was lower and both BRS and HRV were markedly reduced. Zatebradine (0.5 mg/kg i.v.) reduced heart rate in MI-rats from 400 +/- 15 to 350 +/- 19 and in SH-rats from 390 +/- 19 to 324 +/- 6 beats/min without changing mean arterial pressure. Both BRS and HRV were restored in MI- and further increased in SH-rats by the drug. Effects of 0.05, 0.5 and 5 mg/kg zatebradine revealed a dose-dependency of heart rate reduction. The lowest dose enhanced reflex bradycardia despite little effect on heart rate and lack of effect on both reflex tachycardia and HRV. CONCLUSIONS: Both BRS and HRV are reduced in rats early after MI, indicating a depressed reflex and tonic vagal activity. Treatment with zatebradine enhances both BRS and HRV. These data suggest that the drug has both peripheral and central effects, leading to an increase of vagal control of heart rate.


Subject(s)
Baroreflex/drug effects , Benzazepines/therapeutic use , Cardiotonic Agents/therapeutic use , Heart Rate/drug effects , Myocardial Infarction/drug therapy , Analysis of Variance , Animals , Heart/physiopathology , Linear Models , Male , Myocardial Infarction/physiopathology , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric
14.
Hypertension ; 34(6): 1254-8, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10601127

ABSTRACT

The level of renal blood flow (RBF) is controlled by opposing vasoconstrictor and vasodilator influences. In a recent investigation in normotensive dogs, we found that combined blockade of endothelin type A (ET(A)) receptors and angiotensin II formation induces marked increases in RBF that were much larger than the effects of blocking either system alone. The aim of the present study was to determine the contribution of nitric oxide (NO) to this vasodilator response. Experiments were made in 6 conscious, chronically instrumented dogs subjected to 5 different experimental treatments on separate days. Blockade of ET(A) receptors alone by the selective antagonist LU 135252 had only minor effects on RBF compared with time-control experiments. Additional blockade of angiotensin II formation by angiotensin-converting enzyme inhibition with trandolaprilat caused a substantial increase of RBF by approximately 50%. This vasodilation was entirely suppressed when NO formation was prevented by inhibition of NO synthase with N(G)-nitro-L-arginine methyl ester HCl. However, when during NO synthase inhibition renal vascular NO concentrations were clamped at control levels by infusing the NO donor S-nitroso-N-acetyl-D, L-penicillamine, the vasodilator response to combined blockade of ET(A) receptors and angiotensin II formation was completely restored (DeltaRBF approximately 60%). These results indicate that the vasodilation after combined ET(A) receptor blockade and angiotensin-converting enzyme inhibition is not mediated by an increase in NO release but results from the unmasking of the tonic influence that is normally exerted by constitutively released NO. Accordingly, the tonic activity of endothelial NO synthase appears to be of major importance in the physiological regulation of renal vascular resistance by determining the vasomotor responses to endothelin and angiotensin II.


Subject(s)
Angiotensin II/biosynthesis , Endothelin Receptor Antagonists , Nitric Oxide/metabolism , Renal Circulation/physiology , Vasoconstrictor Agents/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Dogs , Drug Interactions , Enzyme Inhibitors/pharmacology , Female , Kidney/blood supply , Kidney/drug effects , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Phenylpropionates/pharmacology , Pyrimidines/pharmacology , Receptor, Endothelin A , Renal Circulation/drug effects , Vasoconstrictor Agents/pharmacology
15.
J Physiol ; 519 Pt 3: 841-50, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10457095

ABSTRACT

1. The transient outward K+ current (Ito) is a major repolarizing ionic current in ventricular myocytes of several mammals. Recently it has been found that its magnitude depends on the origin of the myocyte and is regulated by a number of physiological and pathophysiological signals. 2. The relationship between the magnitude of Ito, action potential duration (APD) and Ca2+ influx (QCa) was studied in rat left ventricular myocytes of endo- and epicardial origin using whole-cell recordings and the action potential voltage-clamp method. 3. Under control conditions, in response to a depolarizing voltage step to +40 mV, Ito averaged 12.1 +/- 2.6 pA pF-1 in endocardial (n = 11) and 24.0 +/- 2.6 pA pF-1 in epicardial myocytes (n = 12; P < 0.01). APD90 (90 % repolarization) was twice as long in endocardial myocytes, whereas QCa inversely depended on the magnitude of Ito. L-type Ca2+ current density was similar in myocytes from both regions. 4. To determine the effects of controlled reductions of Ito on QCa, recordings were repeated in the presence of increasing concentrations of the Ito inhibitor 4-aminopyridine. 5. Inhibition of Ito by as little as 20 % more than doubled QCa in epicardial myocytes, whereas it had only a minor effect on QCa in myocytes of endocardial origin. Further inhibition of Ito led to a progressive increase in QCa in epicardial myocytes; at 90 % inhibition of Ito, QCa was four times larger than the control value. 6. We conclude that moderate changes in the magnitude of Ito strongly affect QCa primarily in epicardial regions. An alteration of Ito might therefore allow for a regional regulation of contractility during physiological and pathophysiological adaptations.


Subject(s)
Calcium/metabolism , Myocardium/metabolism , Potassium Channels/physiology , Action Potentials , Animals , Electrophysiology , Endocardium/cytology , Female , Pericardium/cytology , Rats , Rats, Sprague-Dawley
16.
Am J Physiol ; 277(2): R412-8, 1999 08.
Article in English | MEDLINE | ID: mdl-10444547

ABSTRACT

The aim of the present study was to quantify with a uniform technique the rates of conversion of ANG I to ANG II in the lung and kidney and the degradation of both peptides to biologically inactive products in the pulmonary, renal, and systemic circulation. We infused the peptides intravenously, into the left ventricle, and into the left renal artery of rats and compared their effects on renal blood flow. The measured change in renal blood flow was used as a bioassay parameter to estimate the concentration of circulating ANG II. Mathematical analysis of our data allowed us to calculate conversion and degradation rates. Furthermore, the role of aminopeptidases A (EC 3.4.11.7) and N (EC 3.4.11.2) in the degradation of the peptides in the kidney was investigated by intrarenal infusion of the inhibitor amastatin. Our results show that the conversion rate of ANG I is 75% in the pulmonary and 21% in the renal circulation. Both peptides are degraded by 5% in the pulmonary, by 67% in the systemic, and by 93% in the renal circulation. Amastatin prevented 60% of the renal degradation of the peptides to inactive products, and this effect could be attributed to inhibition of aminopeptidase N. The results indicate that the converting capacity of the kidney is of minor importance for endocrine generation of ANG II but could be useful for the paracrine production.


Subject(s)
Angiotensin II/metabolism , Angiotensin I/metabolism , Kidney/metabolism , Lung/metabolism , Aminopeptidases/metabolism , Angiotensin I/blood , Angiotensin I/pharmacology , Angiotensin II/blood , Angiotensin II/pharmacology , Animals , CD13 Antigens/metabolism , Female , Glutamyl Aminopeptidase , Heart Ventricles , Infusions, Intravenous , Injections, Intra-Arterial , Male , Models, Biological , Pulmonary Circulation/drug effects , Rats , Rats, Wistar , Renal Artery , Renal Circulation/drug effects
17.
Am J Physiol ; 277(1): H1-7, 1999 07.
Article in English | MEDLINE | ID: mdl-10409174

ABSTRACT

Studies in vitro have demonstrated that vasoconstrictor agents increase intracellular Ca(2+) and activate protein kinase C (PKC) to elevate vascular tone. The aim of the present study was to determine the importance of these signaling pathways for angiotensin II (ANG II) and thromboxane A(2) (TxA(2)) in regulating mesenteric blood flow (MBF) in vivo. In anesthetized rats increasing doses of ANG II or the TxA(2) agonist U-46619 were administered into the superior mesenteric artery to reduce MBF. Intra-arterial infusion of inhibitors served to examine the contribution of different pathways: 8-(diethylamino)octyl 3,4,5-trimethoxybenoate hydrochloride (TMB-8) to inhibit intracellular Ca(2+) release, nifedipine to block transmembrane Ca(2+) influx through the L-type Ca(2+) channel, and staurosporine to inhibit PKC. Each of the inhibitors attenuated ANG II-induced reductions in MBF, and all dose-response curves were shifted to the right to an approximately threefold higher ANG II dose. Combinations of the inhibitors revealed that their effects were additive; together they abolished the vasoconstrictor action of ANG II completely. In contrast, the dose-response curve for U-46619 was not affected by any of the inhibitors infused either separately or together. The results demonstrate that a rise in intracellular Ca(2+) and activation of PKC are major mediators of the vasoconstrictor effect of ANG II in mesenteric circulation, but they play a subordinate role, if any, for the effects of TxA(2). Because TxA(2) plays a major role only under pathological conditions, the uncontrolled vasoconstriction appears to be associated with the recruitment of novel signal transduction pathways.


Subject(s)
Angiotensin II/physiology , Mesentery/blood supply , Signal Transduction/physiology , Thromboxane A2/physiology , Vasoconstriction/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Angiotensin II/pharmacology , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Dose-Response Relationship, Drug , Female , Hemodynamics/drug effects , Hemodynamics/physiology , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Wistar , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology
18.
Hypertension ; 33(6): 1420-4, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10373226

ABSTRACT

Previous studies in dogs have shown additive or even synergistic effects of combined angiotensin-converting enzyme inhibition and either nonselective endothelin subtype A/B (ETA/B) or selective endothelin subtype A (ETA) receptor blockade on renal vascular resistance and mean arterial blood pressure. A possible mechanism underlying this interaction may be a stimulation of the renin-angiotensin system during endothelin (ET) receptor blockade. We therefore investigated whether plasma renin activity and renin release are regulated by the ETA receptor. Experiments were made in conscious, chronically instrumented dogs receiving either saline or the selective ETA receptor antagonist LU 135252 (10 mg/kg IV). Eighty to 100 minutes after the administration of LU 135252 (n=5), heart rate (99+/-7 versus 81+/-6 bpm; P<0.05) and renal blood flow (327+/-40 versus 278+/-36 mL/min; P<0.05) were increased significantly, whereas mean arterial blood pressure tended to be lower (93+/-5 versus 105+/-7 mm Hg). These changes were associated with a 2-fold increase in plasma renin activity (0.74+/-0.12 versus 0.37+/-0.10 ng angiotensin I per milliliter per hour; P<0.05). Measurements of renin release at various renal perfusion pressures (n=5) with the use of a vascular occluder implanted around the left renal artery revealed that ETA receptor blockade did not alter renin release at resting renal perfusion pressure (78+/-25 versus 71+/-39 U/min) but strongly enhanced the sensitivity of pressure-dependent renin release <80 mm Hg approximately 2.2-fold. In conclusion, selective ETA receptor blockade is associated with a stimulation of the circulating renin-angiotensin system, which results from both a sensitization of pressure-dependent renin release and a larger proportion of blood pressure values falling into the low pressure range, where renin release is stimulated. These find-ings strengthen the view that ET and the renin-angiotensin system closely interact to regulate vascular resistance and provide a physiological basis for synergistic hypotensive effects of a combined blockade of both pressor systems.


Subject(s)
Endothelin Receptor Antagonists , Hemodynamics/drug effects , Phenylpropionates/pharmacology , Pyrimidines/pharmacology , Renin-Angiotensin System/physiology , Animals , Blood Pressure/drug effects , Dogs , Female , Heart Rate/drug effects , Hemodynamics/physiology , Kidney/blood supply , Male , Receptor, Endothelin A , Renal Circulation/drug effects , Time Factors
19.
Hypertension ; 33(4): 954-60, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10205230

ABSTRACT

In isolated cardiac myocytes, the direct effects of angiotensin II on cellular growth and gene expression were shown to be mediated by endothelin via the endothelin subtype A (ETA) receptor. To determine whether this pathway is also involved in the cardiovascular adaptations to a chronic activation of the renin-angiotensin system in vivo, the effects of a selective ETA receptor antagonist (LU 127043) were investigated in adult rats with renal artery stenosis. Four groups of rats (n=107) were studied over a period of 10 days after surgery: (1) sham-operated animals with saline administration, (2) rats subjected to left renal artery clipping with saline administration, (3) sham-operated rats with LU 127043 administration, and (4) rats subjected to left renal artery clipping with LU 127043 administration. LU 127043 (50 mg/kg) or saline was given by gavage twice daily starting 1 day before the operation. In clipped rats with saline administration, plasma renin activity, the ratio of left ventricular weight to body weight, and mRNAs for beta-myosin heavy chain and atrial natriuretic peptide were significantly elevated as early as 2 days after surgery. Blood pressure started to rise on the third postoperative day and attained a steady state hypertensive level by day 6. Blockade of ETA receptors had no effects on plasma renin activity or the time course of hypertension in clipped animals but completely prevented left ventricular hypertrophy and the re-expression of the beta-myosin heavy chain and atrial natriuretic peptide genes on day 2. While the expressions of the beta-myosin heavy chain and atrial natriuretic peptide genes were not different from saline-treated, clipped animals after day 4, the development of left ventricular hypertrophy remained markedly blunted (-50%) during ETA receptor blockade until day 10. These results show that a continuous blockade of ETA receptors significantly attenuates the development of left ventricular hypertrophy and, more transiently, fetal gene expression in the early phase of renovascular hypertension. Since neither blood pressure nor the increase in plasma renin activity was significantly altered by ETA receptor blockade, the inhibitory influences of the ETA receptor antagonist on left ventricular hypertrophy and gene expression were mediated most likely through a direct blockade of myocardial ETA receptors.


Subject(s)
Blood Pressure/drug effects , Endothelin Receptor Antagonists , Hypertension, Renovascular/complications , Hypertrophy, Left Ventricular/prevention & control , Animals , Atrial Natriuretic Factor/genetics , Female , Heart Rate/drug effects , Myosin Heavy Chains/genetics , Phenylpropionates/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptor, Endothelin A , Receptors, Endothelin/physiology , Renin/blood
20.
Am J Physiol ; 276(3): H1022-7, 1999 03.
Article in English | MEDLINE | ID: mdl-10070087

ABSTRACT

The role of endothelin (ET)-1 in blood pressure homeostasis and the interaction with the renin-angiotensin system (RAS) was investigated in normotensive conscious dogs. ETA receptors were blocked by LU-135252 (1-30 mg/kg); trandolapril (2 mg/kg) or losartan (10 mg/kg) was used to inhibit the RAS. LU-135252 in oral doses of 3-30 mg/kg significantly reduced mean arterial pressure (MAP) by approximately 10 mmHg maximally, whereas trandolapril or losartan were without any effect. MAP reduction was more pronounced when LU-135252 was combined with either losartan (-15.5 +/- 3.2 mmHg; 2 h postadministration; P < 0.05) or trandolapril (-30.9 +/- 3.6 mmHg; P < 0.05). When endogenous nitric oxide (NO) generation was blocked but NO concomitantly infused, this synergistic effect on MAP was prevented. The data show that ET-1 contributes to the maintenance of blood pressure via ETA receptors. Furthermore, ET-1 and ANG II play a prominent role in the control of blood pressure by opposing the effects of NO. The pronounced blood pressure fall after combined blockade of ETA receptors and the RAS may be mediated by an enhanced release of NO.


Subject(s)
Blood Pressure/physiology , Endothelins/physiology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Dogs , Drug Synergism , Endothelin Receptor Antagonists , Indoles/pharmacology , Losartan/pharmacology , Male , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/pharmacology , Phenylpropionates/pharmacology , Pyrimidines/pharmacology , Receptor, Endothelin A , Reference Values
SELECTION OF CITATIONS
SEARCH DETAIL
...