Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Am J Physiol Regul Integr Comp Physiol ; 307(5): R481-3, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25031231

ABSTRACT

The American Physiological Society officially recognized the area of research in Water and Electrolyte Homeostasis (WEH) over 30 years ago when the Section of WEH was established. This minireview illuminates the importance of WEH research to the physiology community. By the narrowest definition, WEH research studies the regulation of body fluids; however, this research area is much broader and more relevant today than when this subdiscipline was first recognized because of the translational and systemic "point of view" of WEH research. This minireview highlights how WEH research serves as a balanced force between the full range of other more traditional organ-based physiological and pathophysiological concepts. The breadth of research in which WEH investigators engage is on full display with the publication of minireviews from the annual Data Diuresis session at Experimental Biology.


Subject(s)
Homeostasis/physiology , Physiological Phenomena/physiology , Water-Electrolyte Balance/physiology , Animals , Body Fluids/physiology , Diuresis/physiology , Electrolytes , Humans
2.
J Am Soc Nephrol ; 19(2): 225-32, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18057218

ABSTRACT

Oxytocin is known to have an antidiuretic effect, but the mechanisms underlying this effect are not completely understood. We infused oxytocin by osmotic minipump into vasopressin-deficient Brattleboro rats for five days and observed marked antidiuresis, increased urine osmolality, and increased solute-free water reabsorption. Administration of oxytocin also significantly increased the protein levels of aquaporin-2 (AQP2), phosphorylated AQP2 (p-AQP2), and AQP3 in the inner medulla and in the outer medulla plus cortex. Immunohistochemistry demonstrated increased AQP2 and p-AQP2 expression and trafficking to the apical plasma membrane of principal cells in the collecting duct, and increased AQP3 expression in the basolateral membrane. These oxytocin-induced effects were blocked by treatment with the vasopressin V2 receptor antagonist SR121463B, but not by treatment with the oxytocin receptor antagonist GW796679X. We conclude that vasopressin V2 receptors mediate the antidiuretic effects of oxytocin, including increased expression and apical trafficking of AQP2, p-AQP2, and increased AQP3 protein expression.


Subject(s)
Diuresis/physiology , Kidney Concentrating Ability/physiology , Kidney Tubules, Collecting/metabolism , Oxytocin/physiology , Animals , Antidiuretic Hormone Receptor Antagonists , Aquaporin 2/metabolism , Aquaporin 3/metabolism , Diuresis/drug effects , Kidney Concentrating Ability/drug effects , Kidney Tubules, Collecting/drug effects , Male , Oxytocin/pharmacology , Phosphorylation/drug effects , Protein Transport/drug effects , Protein Transport/physiology , Rats , Rats, Brattleboro , Receptors, Oxytocin/antagonists & inhibitors , Receptors, Oxytocin/metabolism , Receptors, Vasopressin/metabolism , Water-Electrolyte Balance/drug effects , Water-Electrolyte Balance/physiology
3.
J Immunol Methods ; 336(2): 235-41, 2008 Jul 31.
Article in English | MEDLINE | ID: mdl-18555269

ABSTRACT

Digibind and DigiFab are commercial formulations of polyclonal, ovine, digoxin-specific Fabs in clinical use for treatment of digoxin intoxication. Of interest for extending its use to other clinical indications, Digibind has also been reported to neutralize the effect of endogenous digoxin-like molecules, including ouabain, that are linked to clinical disorders ranging from preeclampsia to congestive heart failure. Although Digibind and DigiFab are equivalent in their digoxin-binding activity, the antigens used to produce these Fabs are different. We therefore explored, using native (3)H-digoxin and (3)H-ouabain in four different types of solution-phase binding methods, whether they might exhibit different profiles with respect to ouabain and other digoxin-like factors. Consistent with previous results, both Fab preparations bound digoxin with the same affinities and capacities. However, (3)H-ouabain was found to bind with high affinity only to Fab sub-populations present in both products. Interestingly, this sub-population was twice as large for Digibind compared to DigiFab. Competition experiments also showed differences in specificity within Fab sub-populations. Therefore, the equivalence in digoxin-binding activity of the two Fab preparations does not extend to ouabain-binding capacity and Fab specificity, with implications for clinical differentiation between the preparations in treatment of disorders related to control of non-digoxin cardenolides. The existence of a small but perhaps clinically relevant sub-population of antibodies was detected using specific radioligands. This sub-population could not have been detected nor quantified using standard cross-reactivity in an ELISA assay.


Subject(s)
Digoxin/immunology , Immunoglobulin Fab Fragments/immunology , Ouabain/immunology , Antibody Specificity , Antigen-Antibody Reactions , Binding Sites , Binding, Competitive , Bufanolides/immunology , Bufanolides/metabolism , Digoxin/metabolism , Immunoglobulin Fab Fragments/metabolism , Ouabain/metabolism
4.
J Pharmacol Exp Ther ; 326(2): 432-42, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18499743

ABSTRACT

The transient receptor potential (TRP) vanilloid 4 (TRPV4) member of the TRP superfamily has recently been implicated in numerous physiological processes. In this study, we describe a small molecule TRPV4 channel activator, (N-((1S)-1-{[4-((2S)-2-{[(2,4-dichlorophenyl)sulfonyl]amino}-3-hydroxypropanoyl)-1-piperazinyl]carbonyl}-3-methylbutyl)-1-benzothiophene-2-carboxamide (GSK1016790A), which we have used as a valuable tool in investigating the role of TRPV4 in the urinary bladder. GSK1016790A elicited Ca2+ influx in mouse and human TRPV4-expressing human embryonic kidney (HEK) cells (EC50 values of 18 and 2.1 nM, respectively), and it evoked a dose-dependent activation of TRPV4 whole-cell currents at concentrations above 1 nM. In contrast, the TRPV4 activator 4alpha-phorbol 12,13-didecanoate (4alpha-PDD) was 300-fold less potent than GSK1016790A in activating TRPV4 currents. TRPV4 mRNA was detected in urinary bladder smooth muscle (UBSM) and urothelium of TRPV4+/+ mouse bladders. Western blotting and immunohistochemistry demonstrated protein expression in both the UBSM and urothelium that was absent in TRPV4-/- bladders. TRPV4 activation with GSK1016790A contracted TRPV4+/+ mouse bladders in vitro, both in the presence and absence of the urothelium, an effect that was undetected in TRPV4-/- bladders. Consistent with the effects on TRPV4 HEK whole-cell currents, 4alpha-PDD demonstrated a weak ability to contract bladder strips compared with GSK1016790A. In vivo, urodynamics in TRPV4+/+ and TRPV4-/- mice revealed an enhanced bladder capacity in the TRPV4-/- mice. Infusion of GSK1016790A into the bladders of TRPV4+/+ mice induced bladder overactivity with no effect in TRPV4-/- mice. Overall TRPV4 plays an important role in urinary bladder function that includes an ability to contract the bladder as a result of the expression of TRPV4 in the UBSM.


Subject(s)
Leucine/analogs & derivatives , Muscle Contraction/drug effects , Sulfonamides/pharmacology , TRPV Cation Channels/agonists , Urinary Bladder/drug effects , Urodynamics/drug effects , Urothelium/drug effects , Animals , Body Weight/drug effects , Female , Leucine/pharmacology , Male , Mice , Mice, Knockout , Molecular Structure , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Phorbols/pharmacology , TRPV Cation Channels/genetics , TRPV Cation Channels/physiology , Urinary Bladder/metabolism , Urothelium/metabolism
6.
Auton Neurosci ; 140(1-2): 53-8, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18495551

ABSTRACT

Genetically targeted animals are used throughout research to investigate the role genes play in biological function, including the lower urinary tract. Generation of transgenic mice involves backcrossing for successive generations. Parental strain background genes can interact with the mutated gene potentially affecting interpretation of the mutant phenotype. Differences in physiological phenotypes may also be influenced by gender. The present study evaluated bladder function in five strains of male and female mice, 129S6/SvEvTac, A/J, B6129F1/Tac, BALB/cAnNCrL and C57BL/6NTacFBr. Urodynamic parameters were analyzed during infusion of saline (threshold and void volume, non-voiding contractions, pressure threshold and bladder contraction amplitude) in conscious mice and using voluntary urination in freely moving mice placed on filter paper (number of small and large diameter urine spots), which represent commonly used techniques in preclinical characterization of bladder function. Female BALB/c mice exhibited a significantly larger number of non-voiding contractions and urine dripping (increased number of small urine spots) compared to other female mice. Male BALB/c mice did not share this phenotype. Significant differences in threshold and void volumes were also noted amongst strains and genders. The numbers of large diameter urine spots differed amongst female, and not male, mouse strains. Gender differences were observed between sexes of the same strain in both large and small urine spots. These data demonstrate that genetic background and gender can influence bladder function in the mouse. These differences have a significant impact on the choice of strain and gender when investigating the effects of genetic manipulation on the micturition reflex.


Subject(s)
Autonomic Nervous System/physiology , Sex Characteristics , Urinary Bladder/physiology , Urination/genetics , Urination/physiology , Animals , Female , Genotype , Male , Mice , Mice, Inbred BALB C/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Muscle Contraction/genetics , Muscle, Smooth/innervation , Muscle, Smooth/physiology , Phenotype , Pressure , Reflex/genetics , Sex Factors , Species Specificity , Urinary Bladder/innervation , Urination Disorders/genetics , Urination Disorders/physiopathology
7.
J Hypertens ; 25(4): 731-41, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17351362

ABSTRACT

Oxidative and inflammatory stresses are cardinal in the pathogenesis of hypertension and atherosclerosis. Oxidative stress also leads to the induction of inflammation through the activation of proinflammatory transcription factors. Understanding the mechanisms leading to oxidative stress and the means of suppressing it are important in controlling complications related to atherogenesis, since oxidative and inflammatory stress are important in the pathogenesis of atherosclerosis. The failure of chemical antioxidants [which scavenge reactive oxygen species (ROS)], such as vitamins E and C, has led to further exploration of the ROS-suppressive effects of drugs used in the treatment of cardiovascular disease. Carvedilol has been shown to possess both ROS-scavenging and ROS-suppressive effects, and its use is associated with a reduction in oxidative stress. Furthermore, anti-inflammatory effects of carvedilol have now been described. Although further clinical investigations are required, these properties may contribute to the improvement in clinical outcomes observed with carvedilol.


Subject(s)
Antioxidants/pharmacology , Carbazoles/pharmacology , Cardiovascular Diseases/metabolism , Propanolamines/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Cardiovascular Diseases/physiopathology , Carvedilol , Humans , Myocardial Contraction/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
8.
Eur J Pharmacol ; 563(1-3): 203-8, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17382925

ABSTRACT

Acetic acid was found to have actions on urinary bladder smooth muscle in our routine ion channel screening assays. Numerous studies have examined the mechanisms of bladder irritation by acetic acid; however, the direct effect of acetic acid on ion channels in detrusor smooth muscle cells has not been evaluated. We used whole-cell patch-clamp techniques to examine the effect of acetic acid on large-conductance Ca2+-activated K+ channels (BKCa) from guinea pig detrusor smooth muscle cells and CHO cells expressing recombinant human BKCaalphabeta1 (CHO BKCaalphabeta1) and human BKCaalpha (CHO BKCaalpha). Acetic acid activated BKCa currents in a concentration-dependent (0.01% to 0.05% v/v) manner in all the cell systems studied. Acetic acid (0.05%) increased BKCa current at +30 mV by 2764+/-918% (n=8) in guinea pig detrusor smooth muscle cells. Acetic acid (0.03%) shifted the V1/2 of conductance-voltage curve by 64+/-14 (n=5), 128+/-14 (n=5), and 126+/-12 mV (n=4) in CHO BKCaalpha, CHO BKCaalphabeta1 and detrusor smooth muscle cells, respectively. This effect of acetic acid was found to be independent of pH and was also not produced by its salt form, sodium acetate. Automated patch-clamp experiments also showed similar activation of CHO BKCaalphabeta1 by acetic acid. In conclusion, acetic acid directly activates BKCa channels in detrusor smooth muscle cells. This novel study necessitates caution while interpreting the results from acetic acid bladder irritation model.


Subject(s)
Acetic Acid/pharmacology , Irritants/pharmacology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/agonists , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/agonists , Myocytes, Smooth Muscle/drug effects , Urinary Bladder/drug effects , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Guinea Pigs , Humans , In Vitro Techniques , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Membrane Potentials/drug effects , Myocytes, Smooth Muscle/metabolism , Patch-Clamp Techniques , Recombinant Proteins/agonists , Transfection , Urinary Bladder/cytology , Urinary Bladder/metabolism
9.
Br J Pharmacol ; 145(5): 620-35, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15852036

ABSTRACT

1. SB-706375 potently inhibited [(125)I]hU-II binding to both mammalian recombinant and 'native' UT receptors (K(i) 4.7+/-1.5 to 20.7+/-3.6 nM at rodent, feline and primate recombinant UT receptors and K(i) 5.4+/-0.4 nM at the endogenous UT receptor in SJRH30 cells). 2. Prior exposure to SB-706375 (1 microM, 30 min) did not alter [(125)I]hU-II binding affinity or density in recombinant cells (K(D) 3.1+/-0.4 vs 5.8+/-0.9 nM and B(max) 3.1+/-1.0 vs 2.8+/-0.8 pmol mg(-1)) consistent with a reversible mode of action. 3. The novel, nonpeptidic radioligand [(3)H]SB-657510, a close analogue of SB-706375, bound to the monkey UT receptor (K(D) 2.6+/-0.4 nM, B(max) 0.86+/-0.12 pmol mg(-1)) in a manner that was inhibited by both U-II isopeptides and SB-706375 (K(i) 4.6+/-1.4 to 17.6+/-5.4 nM) consistent with the sulphonamides and native U-II ligands sharing a common UT receptor binding domain. 4. SB-706375 was a potent, competitive hU-II antagonist across species with pK(b) 7.29-8.00 in HEK293-UT receptor cells (inhibition of [Ca(2+)](i)-mobilization) and pK(b) 7.47 in rat isolated aorta (inhibition of contraction). SB-706375 also reversed tone established in the rat aorta by prior exposure to hU-II (K(app) approximately 20 nM). 5. SB-706375 was a selective U-II antagonist with >/=100-fold selectivity for the human UT receptor compared to 86 distinct receptors, ion channels, enzymes, transporters and nuclear hormones (K(i)/IC(50)>1 microM). Accordingly, the contractile responses induced in isolated aortae by KCl, phenylephrine, angiotensin II and endothelin-1 were unaltered by SB-706375 (1 microM). 6. In summary, SB-706375 is a high-affinity, surmountable, reversible and selective nonpeptide UT receptor antagonist with cross-species activity that will assist in delineating the pathophysiological actions of U-II in mammals.


Subject(s)
Pyrrolidines/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Sulfonamides/pharmacology , Algorithms , Animals , Aorta, Thoracic/drug effects , Binding, Competitive/drug effects , Cats , Cell Line, Tumor , Cell Membrane/metabolism , Haplorhini , Humans , In Vitro Techniques , Mice , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Radioligand Assay , Rats , Recombinant Proteins/metabolism , Rhabdomyosarcoma/metabolism , Species Specificity
10.
Br J Pharmacol ; 139(8): 1383-8, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12922924

ABSTRACT

1. Loop and thiazide diuretics are common therapeutic agents for the treatment of sodium retention and oedema. However, resistance to diuretics and decreases in renal function can develop during diuretic therapy. Adenosine causes renal vasoconstriction, sodium reabsorption, and participates in the tubuloglomerular feedback mechanism for the regulation of glomerular filtration rate. 2. We tested the hypothesis that the selective adenosine A(1) receptor antagonist FK838 is orally active and causes diuresis and natriuresis, but maintains glomerular filtration rate in normal rats or in rats with furosemide resistance. 3. In normal male Sprague - Dawley rats, FK838 dose-dependently increased urine flow and sodium and chloride excretion while sparing potassium. In combination with furosemide, FK838 enhanced the diuretic and natriuretic actions of furosemide to the same extent as hydrochlorothiazide and did not increase the potassium loss in normal rats. In furosemide-resistant rats, FK838 increased urine flow and electrolyte excretion to a greater extent than hydrochlorothiazide. In addition, hydrochlorothiazide significantly decreased glomerular filtration rate, whereas FK838 maintained glomerular filtration rate in furosemide-resistant rats. 4. This study shows that the adenosine A(1) receptor antagonist FK838 is orally active and causes potent diuresis and natriuresis and maintains glomerular filtration rate in normal or furosemide-resistant rats. Adenosine A(1) receptor antagonists may be novel therapeutics for the treatment of oedema in normal or otherwise diuretic-resistant patients.


Subject(s)
Adenosine A1 Receptor Antagonists , Diuresis/drug effects , Diuretics/pharmacology , Furosemide/pharmacology , Glomerular Filtration Rate/drug effects , Pyrazoles/pharmacology , Pyridines/pharmacology , Administration, Oral , Animals , Chlorides/urine , Dose-Response Relationship, Drug , Drug Synergism , Hydrochlorothiazide/pharmacology , Male , Potassium/urine , Rats , Rats, Sprague-Dawley , Sodium/urine
12.
Pharmacology ; 80(2-3): 166-76, 2007.
Article in English | MEDLINE | ID: mdl-17551266

ABSTRACT

BACKGROUND/AIMS: Histological studies have provided evidence that carvedilol can prevent cardiac hypertrophy in spontaneously hypertensive-stroke prone rats (SP) fed a high-fat and -salt diet. However, the effects of carvedilol on cardiac function have not been studied in these animals. In addition, the ability of carvedilol to reverse established cardiac hypertrophy and dysfunction under these conditions remains to be determined. Here we have evaluated the ability of carvedilol to prevent and reverse cardiac hypertrophy and progressive dysfunction using echocardiography. METHODS: Two echocardiology studies were conducted to determine the effects of carvedilol treatment on cardiac hypertrophy and dysfunction. In the first prevention study, four groups of rats were evaluated. SP were fed a high-fat (24.5% in food) and high-salt (1% in water) diet (SFD) without (SP-SFD control group) or with carvedilol (SP-SFD carvedilol group; carvedilol concentration 2,400 parts per million) for 18 weeks. Carvedilol was administered in the food at an optimum concentration (i.e. known to provide clinically relevant blood concentrations and reduce cardiac hypertrophy determined from previous studies). In addition, SP and WKY rats were fed a normal diet (SP normal diet group and WKY normal diet group). These groups are known to not develop the same significant cardiac hypertrophy and dysfunction within this limited time of study, and provided two more normal control groups for comparison. In the second reversal study, one group of SP was fed SFD for 12 weeks (SP-SFD pretreatment period) to induce cardiac hypertrophy. Carvedilol (2,400 parts per million) was then added to the diet for an additional 6 weeks (SP-SFD carvedilol treatment period). RESULTS: In the first prevention study, carvedilol prolonged longevity (p < 0.05) and prevented left-ventricular hypertrophy and dysfunction (p < 0.05; SP-SFD control vs. SP-SFD carvedilol group). M-mode-measured and -calculated parameters demonstrated that carvedilol treatment in the SP-SFD carvedilol group prevented increases in left-ventricular wall thickness (p < 0.05) and decreases in diastolic chamber diameter and volume, stroke volume, ejection fraction and cardiac output (all p < 0.05) that occurred in the SP-SFD control group. Further, cardiac measurements in the SP-SFD carvedilol group were normalized to levels similar to those in the SP and WKY normal diet groups. All SFD-fed groups exhibited similar, significantly elevated blood pressure during the study. In the second reversal study, carvedilol treatment for 6 weeks reversed the cardiac hypertrophy and dysfunction that developed in SP-fed SFD for 12 weeks prior to carvedilol intervention. Under these conditions, carvedilol improved/normalized left-ventricular wall thickness, diastolic ventricular-chamber diameter and volume, stroke volume, ejection fraction and cardiac output (all p < 0.05). CONCLUSIONS: These data indicate that carvedilol provides protection from and facilitates reversal of progressive cardiac remodeling and dysfunction in this SP-SFD model of cardiac hypertrophy/heart failure. Since these effects occurred in the absence of effects on blood pressure, other known actions of carvedilol, especially its antioxidant activity, for example, may explain this significant cardiac protection. In addition, research using this SP-SFD model of cardiac hypertrophy/end-organ injury appears to translate well to human cardiovascular disease.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Antioxidants/pharmacology , Carbazoles/pharmacology , Cardiomegaly/prevention & control , Propanolamines/pharmacology , Vasodilator Agents/pharmacology , Administration, Oral , Animals , Cardiomegaly/diagnostic imaging , Cardiomegaly/physiopathology , Carvedilol , Diet , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Treatment Outcome , Ultrasonography , Ventricular Remodeling/drug effects
13.
Fertil Steril ; 88(4 Suppl): 1058-64, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17362941

ABSTRACT

OBJECTIVE: To non-invasively characterize ectopic uterine tissue (EUT) development in a modified autologous rat surgical model of endometriosis using magnetic resonance imaging (MRI). DESIGN: Investigational MRI study. SETTING: A pharmaceutical company. ANIMAL(S): Female Sprague Dawley rats. INTERVENTION(S): Uterine tissue was autotransplanted on the right peritoneal wall of rats. Rats were serially imaged after surgery and after endogenous hormone suppression, hormone supplementation, or ovariectomy. In addition, an MRI contrast agent was administered to examine EUT perfusion characteristics. MAIN OUTCOME MEASURE(S): Changes in transplanted EUT volume and perfusion were monitored using MRI. RESULT(S): The EUT growth could be readily monitored non-invasively by MRI. Although EUT growth was rapid during the initial 4 days after surgery, volume stabilized by the third week and maintained for at least 9 weeks after transplantation. The EUT volumes varied with the estrous cycle and were hormonally sensitive to ovariectomy, to Antide (GnRH antagonist), and to Antide followed by 17beta-E(2) supplementation. The use of an MRI contrast agent facilitated visualization of EUT wall perfusion. CONCLUSION(S): MRI allows for noninvasive, dynamic evaluation of transplanted EUT growth in the rat. This reproducible model will allow for performing quantifiable pharmacologic studies in pre-clinical drug discovery for therapies targeting endometriosis.


Subject(s)
Choristoma/diagnosis , Magnetic Resonance Imaging/methods , Peritoneal Cavity , Uterus , Animals , Choristoma/physiopathology , Endometriosis/diagnosis , Endometriosis/physiopathology , Estrous Cycle/physiology , Female , Rats , Rats, Sprague-Dawley
14.
Am J Physiol Regul Integr Comp Physiol ; 293(1): R299-305, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17395790

ABSTRACT

Spontaneous and induced uterine contractions in the rat were found to be inhibited by a novel and selective oxytocin receptor antagonist GSK221149A (3R,6R)-3-Indan-2-yl-1-[(1R)-1-(2-methyl-1,3-oxazol-4-yl)-2-morpholin-4-yl-2-oxoethyl]-6-[(1S)-1-methylpropyl]-2,5-piperazinedione. GSK221149A displayed nanomolar affinity (K(i) = 0.65 nM) for human recombinant oxytocin receptors with >1,400-fold selectivity over human V1a, V1b, and V2 receptors. GSK221149A had similar affinity (K(i) = 4.1 nM) and selectivity for native oxytocin receptors from rat and produced a functional, competitive block of oxytocin-induced contractions in isolated rat myometrial strips with a pA(2) value of 8.18. Intravenous administration of GSK221149A produced a dose-dependent decrease in oxytocin-induced uterine contractions in anesthetized rats with an ID(50) = 0.27 +/- 0.60 mg/kg (corresponding plasma concentrations were 88 ng/ml). Oral administration of GSK221149A (5 mg/kg) was effective in inhibiting oxytocin-induced uterine contractions after single and multiple (4-day) dosing. Spontaneous uterine contractions in late-term pregnant rats (19-21 days gestation) were significantly reduced by intravenous administration of 0.3 mg/kg of GSK221149A. These results provide further evidence that selective oxytocin receptor antagonism may offer an effective treatment for preterm labor.


Subject(s)
Oxytocin/antagonists & inhibitors , Oxytocin/pharmacology , Piperazines/pharmacology , Receptors, Oxytocin/antagonists & inhibitors , Uterine Contraction/physiology , Anesthesia , Animals , Binding, Competitive/drug effects , CHO Cells , Cell Line , Cricetinae , Cricetulus , Female , Humans , Parity , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Vasopressin/drug effects , Transfection , Vasopressins/pharmacology
15.
J Pharmacol Exp Ther ; 319(2): 957-62, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16928897

ABSTRACT

Human ether-a-go-go-related gene (hERG) encodes a rapidly activating delayed rectifier potassium channel that plays important roles in cardiac action potential repolarization. Although many drugs and compounds block hERG channels, activators of the channel have only recently been described. Three structurally diverse synthetic compounds have been reported to activate hERG channels by altering deactivation or inactivation or by unidentified mechanisms. Here, we describe a novel, naturally occurring hERG channel activator, mallotoxin (MTX). The effects of MTX on hERG channels were investigated using the patch-clamp technique. MTX increased both step and tail hERG currents with EC(50) values of 0.34 and 0.52 microM, respectively. MTX leftward shifted the voltage dependence of hERG channel activation to less depolarized voltages ( approximately 24 mV at 2.5 microM). In addition, MTX increased hERG deactivation time constants. MTX did not change the half-maximal inactivation voltage of the hERG channel, but it reduced the slope of the voltage-dependent inactivation curve. All of these factors contribute to the enhanced activity of hERG channels. During a voltage-clamp protocol using prerecorded cardiac action potentials, 2.5 microM MTX increased the total potassium ions passed through hERG channels by approximately 5-fold. In conclusion, MTX activates hERG channels through distinct mechanisms and with significantly higher potency than previously reported hERG channel activators.


Subject(s)
Acetophenones/pharmacology , Benzopyrans/pharmacology , Enzyme Inhibitors/pharmacology , Ether-A-Go-Go Potassium Channels/drug effects , Action Potentials/drug effects , Animals , CHO Cells , Cricetinae , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/physiology , Potassium/metabolism , Protein Kinase C/antagonists & inhibitors , Rabbits
16.
J Pharmacol Exp Ther ; 313(3): 943-51, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15769863

ABSTRACT

SB-525334 (6-[2-tert-butyl-5-(6-methyl-pyridin-2-yl)-1H-imidazol-4-yl]-quinoxaline) has been characterized as a potent and selective inhibitor of the transforming growth factor-beta1 (TGF-beta1) receptor, activin receptor-like kinase (ALK5). The compound inhibited ALK5 kinase activity with an IC(50) of 14.3 nM and was approximately 4-fold less potent as an inhibitor of ALK4 (IC(50) = 58.5 nM). SB-525334 was inactive as an inhibitor of ALK2, ALK3, and ALK6 (IC(50) > 10,000 nM). In cell-based assays, SB-525334 (1 microM) blocked TGF-beta1-induced phosphorylation and nuclear translocation of Smad2/3 in renal proximal tubule cells and inhibited TGF-beta1-induced increases in plasminogen activator inhibitor-1 (PAI-1) and procollagen alpha1(I) mRNA expression in A498 renal epithelial carcinoma cells. In view of this profile, SB-525334 was used to investigate the role of TGF-beta1 in the acute puromycin aminonucleoside (PAN) rat model of renal disease, a model of nephritis-induced renal fibrosis. Orally administered doses of 1, 3, or 10 mg/kg/day SB-525334 for 11 days produced statistically significant reductions in renal PAI-1 mRNA. Also, the compound produced dose-dependent decreases in renal procollagen alpha1(I) and procollagen alpha1(III) mRNA, which reached statistical significance at the 10-mg/kg/day dose when compared with vehicle-treated PAN controls. Furthermore, PAN-induced proteinuria was significantly inhibited at the 10-mg/kg/day dose level. These results provide further evidence for the involvement of TGF-beta1 in the profibrotic changes that occur in the PAN model and for the first time, demonstrate the ability of a small molecule inhibitor of ALK5 to block several of the markers that are predictive of fibrosis and renal injury in this model.


Subject(s)
Kidney/pathology , Nephritis/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Puromycin/toxicity , Transforming Growth Factor beta/antagonists & inhibitors , Activin Receptors/antagonists & inhibitors , Animals , Biomarkers , Cell Line, Tumor , Collagen Type I/genetics , Dose-Response Relationship, Drug , Fibrosis , Humans , Nephritis/metabolism , Phosphorylation , Plasminogen Activator Inhibitor 1/genetics , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta , Transforming Growth Factor beta/physiology , Transforming Growth Factor beta1
17.
J Pharmacol Exp Ther ; 315(3): 1306-13, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16144980

ABSTRACT

We describe N-[(2S)-2-(mercaptomethyl)-3-methylbutanoyl]-4-(1H-pyrazol-1-yl)-L-phenylalanine (GW796406), a vasopeptidase inhibitor (VPI) that possessed approximately 3-fold selectivity for neutral endopeptidase 24.11 (NEP) versus angiotensin-converting enzyme (ACE) in in vitro assays using rat and human enzymes. In the same assays, omapatrilat, the most extensively studied VPI, displayed approximately 3-fold selectivity for ACE. The in vivo ACE and NEP inhibition profile and the liability of the compounds to increase plasma extravasation were compared at two (low and high) therapeutically equivalent intravenous doses in the rat. At the low dose, both agents inhibited ACE activity by approximately 85%. Consistent with their in vitro ACE/NEP selectivity, omapatrilat produced 49% inhibition, whereas GW796406 produced >95% inhibition of NEP. Neither compound increased plasma extravasation. When the low dose was administered to rats pretreated with the NEP inhibitor ecadotril to normalize NEP background to <5% of control, only omapatrilat significantly increased plasma extravasation. At the high dose, omapatrilat and GW796406 produced profound, nonselective inhibition of ACE (>90%) and NEP (>95%), and they significantly increased plasma extravasation. The activity of the agents as inhibitors of dipeptidylpeptidase IV (DPP IV) and aminopeptidase P (APP) was also investigated. Neither compound inhibited DPP IV. Interestingly, omapatrilat, but not GW796406, was a relatively potent inhibitor of APP (IC50 = 260 nM). We investigated whether APP inhibition increased the plasma extravasation liability of GW796406. The low dose of GW796406 administered with apstatin, an APP inhibitor, did not increase plasma extravasation. This finding inferred that APP inhibition is not involved in plasma extravasation in the rat and that APP inhibition does not explain the increased plasma extravasation produced by omapatrilat in NEP-inhibited rats.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Neprilysin/pharmacology , Plasma/drug effects , Pyridines/pharmacology , Thiazepines/pharmacology , Aminopeptidases/analysis , Aminopeptidases/antagonists & inhibitors , Angiotensin-Converting Enzyme Inhibitors/analysis , Animals , Dipeptidyl Peptidase 4/analysis , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Kidney/drug effects , Kidney/enzymology , Lung/drug effects , Lung/enzymology , Male , Neprilysin/analysis , Peptides/pharmacology , Phenylalanine/analogs & derivatives , Phenylalanine/chemistry , Phenylalanine/pharmacology , Plasma/physiology , Pyrazoles/chemistry , Pyrazoles/pharmacology , Rabbits , Rats , Rats, Sprague-Dawley , Rats, Wistar
18.
J Pharmacol Exp Ther ; 310(1): 319-25, 2004 Jul.
Article in English | MEDLINE | ID: mdl-14982968

ABSTRACT

Digoxin-specific Fab (Digibind) is a mixture of antidigoxin Fab fragments prepared from sheep sera and is used as a treatment for digoxin poisoning. Digoxin-specific Fab has been shown to neutralize an endogenous Na+/K+ ATPase inhibitor (endogenous digoxin-like Na+/K+ ATPase regulatory factor; EDLF) in rats and humans and to lower blood pressure. Although the exact structure of EDLF is unknown, compounds identical to or structurally related to ouabain, bufalin, and marinobufagenin have been detected in mammalian plasma. In this study, some structural characteristics of EDLF were inferred from the ability of digoxin-specific Fab to neutralize the Na+/K+ ATPase inhibitory activity of several known cardenolides and bufodienolides. Additional structural information was obtained from [3H]ouabain binding and enzyme-linked immunosorbent assay experiments. Digoxin-specific Fab had the ability to interact to some extent with all of the cardenolides and bufodienolides tested. However, digoxin-specific Fab was more than 20-fold more potent in neutralizing ouabain and bufalin than marinobufagenin. The antihypertensive effect of digoxin-specific Fab seen in preeclampsia and animal models of hypertension may therefore be due to a molecule identical to or structurally similar to ouabain or bufalin.


Subject(s)
Immunoglobulin Fab Fragments/metabolism , Ouabain/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Digoxin/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Hypertension/metabolism , Rats , Steroids/metabolism , Tritium
19.
J Pharmacol Exp Ther ; 309(3): 1141-7, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14769834

ABSTRACT

The effect of angiotensin-converting enzyme (ACE) and neutral endopeptidase (NEP) inhibition on microvascular plasma leakage (extravasation) was evaluated in a rat model. Progressive inhibition of ACE using captopril caused increased extravasation when lung ACE was inhibited by >55%. In contrast, the selective inhibition of renal NEP by >90% using ecadotril did not increase extravasation. In NEP-inhibited rats, extravasation produced by the ACE inhibitors captopril and lisinopril was markedly enhanced. The dual ACE and NEP inhibitor omapatrilat, at oral doses of 0.03, 0.1, and 0.3 mg/kg, selectively inhibited lung ACE by 19, 61, and 76%, respectively, and did not cause significant extravasation. Doses of 1 and 10 mg/kg omapatrilat, which produced >90% inhibition of ACE and also inhibited renal NEP by 54 and 78%, respectively, significantly increased extravasation. In this model, bradykinin and substance P produced extravasation that could be abolished by the bradykinin 2 (B2) receptor antagonist Hoe 140 (icatibant) or the neurokinin1 (NK1) antagonist CP99994 [(+)-(2S,3S)-3-(2-methoxybenzylamino)-2-phenylpiperidine], respectively. Bradykinin induced extravasation was also partially ( approximately 40%) inhibited by CP99994, indicating that a portion of the response involves B2 receptor-mediated release of substance P. In conclusion, this study is the first to relate the degree of ACE and/or NEP inhibition to extravasation liability in the rat model. Our data clearly demonstrate that ACE inhibitor-induced plasma extravasation is enhanced by concomitant inhibition of NEP. In addition, this study provides further evidence for the role for B2 and NK1 receptors in mediating plasma extravasation in the rat.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Bradykinin/analogs & derivatives , Captopril/pharmacology , Neprilysin/antagonists & inhibitors , Peptidyl-Dipeptidase A/metabolism , Plasma/drug effects , Thiorphan/analogs & derivatives , Animals , Bradykinin/pharmacology , Dose-Response Relationship, Drug , Lung/drug effects , Lung/enzymology , Male , Plasma/physiology , Rats , Rats, Sprague-Dawley , Receptor, Bradykinin B1/drug effects , Receptor, Bradykinin B1/physiology , Receptor, Bradykinin B2/drug effects , Receptor, Bradykinin B2/physiology , Receptors, Neurokinin-1/drug effects , Receptors, Neurokinin-1/physiology , Substance P/pharmacology , Thiorphan/pharmacology
20.
Mol Cell Biochem ; 260(1-2): 7-11, 2004 May.
Article in English | MEDLINE | ID: mdl-15228080

ABSTRACT

Glomerular mesangial cells play an important role in the development of glomerulosclerosis. Mesangial cell apoptosis has been shown to be involved in different stages of development of glomerulonephritis. The aim of the present study was to evaluate the effect of inhibition of serine/threonine phosphatases by okadaic acid, a shell fish toxin, on rat mesangial cell apoptosis and to examine the molecular mechanisms particularly the role of caspases. Okadaic acid significantly induced mesangial cell apoptosis, as measured by an increase in cytoplasmic nucleosome-associated DNA fragmentation. The induction of apoptosis was dependent on protein synthesis, because cyclohexamide, a protein synthesis inhibitor, blocked okadaic acid-induced apoptosis. In addition, okadaic acid stimulated caspase activities (as measured by caspase substrate peptide hydrolysis) in cultured rat mesangial cells at different time points. After 12 h treatment, okadaic acid caused a modest increase in caspase-8 (IETD-pNAse) (159.3 +/- 6.7%) activity, while after 18 h treatment, okadaic acid caused a significant increase in caspase-3 (DEVD-pNAse) (906 +/- 245%) activity. Okadaic acid-stimulated caspase-3 activity was inhibited by Z-IETD-FMK (caspase-8 inhibitor) suggesting that the caspase-3 activity is downstream of caspase-8 activity. Both caspase-3 and caspase-8 inhibitors blocked okadaic acid-stimulated apoptosis. These data suggest that inhibition of protein phosphatases by okadaic acid induces apoptosis in rat mesangial cells by activating caspase-3- and -8-like activities and that caspase-3-like activity is downstream of caspase-8-like activity.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Glomerular Mesangium/drug effects , Okadaic Acid/pharmacology , Animals , Caspase 3 , Caspase 8 , Caspase Inhibitors , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Glomerular Mesangium/cytology , Glomerular Mesangium/metabolism , Hydrolysis/drug effects , Oligopeptides/pharmacology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL