Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 249
Filter
1.
J Pharmacol Exp Ther ; 298(1): 219-25, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11408545

ABSTRACT

The recombinant alpha2-adrenoceptors, designated as alpha2a and alpha2d, have highly similar amino acid sequences, but distinct pharmacological properties. It has been suggested that these two receptor subtypes are species orthologs, since the alpha2-adrenoceptors of a given species have pharmacological characteristics corresponding to either the alpha2a- (human, pig) or alpha2d- (rat, mouse, guinea pig, cow) adrenoceptor. Radioligand binding assays in rabbit adipocyte suggest alpha2D-adrenoceptor pharmacology. However, functional studies examining prejunctional alpha2-adrenoceptors in several tissues pharmacologically define the receptor of the rabbit as an alpha2A-adrenoceptor rather than an alpha2D-adrenoceptor. We characterized the alpha2-adrenoceptor of rabbit adipocyte and platelet, comparing the ability of norepinephrine and 13 adrenoceptor antagonists to inhibit the binding of [3H]RX821002 with the affinity of these drugs for the human alpha2a-adrenoceptor or the rat alpha2d-adrenoceptor. Pharmacological characteristics of the adipocyte and platelet receptor were very similar, with an excellent correlation between pK(i) values (r2 = 0.95, slope of regression = 1.01). Drug affinities for both platelet and adipocyte receptors correlated better with the alpha2a-adrenoceptor (r2 = 0.68-0.77) than with the alpha2d-adrenoceptor (r2 = 0.37-0.38). Despite the relatively low affinity of the rabbit adipocyte alpha2-adrenoceptor for yohimbine and rauwolscine, this receptor, as well as the platelet receptor, have alpha2A-adrenoceptor pharmacology. Subtle differences in the alpha2-adrenoceptor binding characteristics of these native rabbit tissues compared with the recombinant human alpha2a-adrenoceptor may result either from minor differences in the sequence of human and rabbit alpha2a-adrenoceptors or from differences in the environment to which native and recombinant receptors are exposed.


Subject(s)
Adipocytes/metabolism , Adrenergic alpha-Antagonists/metabolism , Blood Platelets/metabolism , Idazoxan/metabolism , Norepinephrine/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Animals , Idazoxan/analogs & derivatives , Rabbits , Radioligand Assay , Species Specificity
2.
J Virol ; 75(6): 2792-802, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11222703

ABSTRACT

The prototype JHM strain of murine hepatitis virus (MHV) is an enveloped, RNA-containing coronavirus that has been selected in vivo for extreme neurovirulence. This virus encodes spike (S) glycoproteins that are extraordinarily effective mediators of intercellular membrane fusion, unique in their ability to initiate fusion even without prior interaction with the primary MHV receptor, a murine carcinoembryonic antigen-related cell adhesion molecule (CEACAM). In considering the possible role of this hyperactive membrane fusion activity in neurovirulence, we discovered that the growth of JHM in tissue culture selected for variants that had lost murine CEACAM-independent fusion activity. Among the collection of variants, mutations were identified in regions encoding both the receptor-binding (S1) and fusion-inducing (S2) subunits of the spike protein. Each mutation was separately introduced into cDNA encoding the prototype JHM spike, and the set of cDNAs was expressed using vaccinia virus vectors. The variant spikes were similar to that of JHM in their assembly into oligomers, their proteolysis into S1 and S2 cleavage products, their transport to cell surfaces, and their affinity for a soluble form of murine CEACAM. However, these tissue culture-adapted spikes were significantly stabilized as S1-S2 heteromers, and their entirely CEACAM-dependent fusion activity was delayed or reduced relative to prototype JHM spikes. The mutations that we have identified therefore point to regions of the S protein that specifically regulate the membrane fusion reaction. We suggest that cultured cells, unlike certain in vivo environments, select for S proteins with delayed, CEACAM-dependent fusion activities that may increase the likelihood of virus internalization prior to the irreversible uncoating process.


Subject(s)
Genetic Variation , Glycoproteins/metabolism , Membrane Fusion/physiology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Murine hepatitis virus/pathogenicity , Receptors, Virus/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/physiology , Animals , Antigens, CD , Carcinoembryonic Antigen/metabolism , Cell Adhesion Molecules/metabolism , Cell Line , Coronavirus Infections/virology , Gene Expression Regulation, Viral , Giant Cells , Humans , Membrane Glycoproteins/chemistry , Mice , Murine hepatitis virus/physiology , Mutation , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/chemistry
3.
Mol Interv ; 1(5): 250-6, 2001 Dec.
Article in English | MEDLINE | ID: mdl-14993364

ABSTRACT

By his own admission, Robert R. Ruffolo, Head of Research and Development at Wyeth-Ayerst, is a bit of a nerd. Opting to spend seven nights per week with his textbooks at the expense of all else, he earned his pharmacy degree summa cum laude, and his PhD in pharmacology in just over three years. He speaks with unabashed enthusiasm for the pharmaceutical industry, for biomedicine, and particularly for the future of pharmacology. Even if you don't know Ruffolo, you've probably seen him-if not at a science symposium, then surely as the lead "actor" in televised promotions that ran throughout 1999 on behalf of the Pharmaceutical Research and Manufacturers of America (PhRMA; scenes reproduced here, with permission). "Actor" belongs in quotation marks, because if there is such a thing as type casting, then Ruffolo is certainly an example in the PhRMA ads: passionate about good drugs and the people who need them; proud of his profession and his contributions; dedicated to science as well as to his colleagues; and grateful for the opportunities he has had to contribute. The commercial's requisite happy ending, where three generations of a coronary-prone farm family ride off into the sunset, reflects Ruffolo's own success story in helping to bring carvedilol to market. In all of this, however, Ruffolo's sincerity transcends the hokey as well as the nerdy. His devotion to science includes a mission to help others, and he would argue that in this and most ways, he is not so different from his academic colleagues.


Subject(s)
Pharmacology/history , History, 20th Century , History, 21st Century
4.
Eur J Pharmacol ; 406(1): 109-16, 2000 Oct 06.
Article in English | MEDLINE | ID: mdl-11011041

ABSTRACT

Carvedilol, a selective alpha(1) and non-selective beta-adrenoceptor antagonist and antioxidant, has been shown to provide significant cardiac protection in animal models of myocardial ischemia. To further explore the mechanisms contributing to the efficacy of carvedilol cardioprotection, the effects of carvedilol on hemodynamic variables, infarct size and myeloperoxidase activity (an index of neutrophil accumulation) were compared with a beta(1) selective adrenoceptor antagonist, bisoprolol. Carvedilol (1 mg/kg) or bisoprolol (1 mg/kg) was given intravenously 5 min before reperfusion. In vehicle-treated rabbits, ischemia (45 min) and reperfusion (240 min) resulted in significant increases in left ventricular end diastolic pressure, large myocardial infarction (64.7+/-2.6% of area-at-risk) and a marked increase in myeloperoxidase activity (64+/-14 U/g protein in area-at-risk). Carvedilol treatment resulted in sustained reduction of the pressure-rate-index and significantly smaller infarcts (30+/-2.9, P<0.01 vs. vehicle) as well as decreased myeloperoxidase activity (26+/-11 U/g protein in area-at-risk, P<0.01 vs. vehicle). Administration of bisoprolol at 1 mg/kg resulted in a pressure-rate-index comparable to that of carvedilol and also decreased infarct size (48.4+/-2.5%, P<0.001 vs. vehicle, P<0.05 vs. carvedilol), although to a significantly lesser extent than that observed with carvedilol. Treatment with bisoprolol failed to reduce myeloperoxidase activity in the ischemic myocardial tissue. In addition, carvedilol, but not bisoprolol, markedly decreased cardiac membrane lipid peroxidation measured by thiobarbituric acid formation. Taken together, this study suggests that the superior cardioprotection of carvedilol over bisoprolol is possibly the result of carvedilol's antioxidant and anti-neutrophil effects, not its hemodynamic properties.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Bisoprolol/pharmacology , Carbazoles/pharmacology , Cardiovascular Agents/pharmacology , Propanolamines/pharmacology , Reperfusion Injury/prevention & control , Adrenergic beta-Agonists/pharmacology , Animals , Antioxidants/pharmacology , Carvedilol , Creatine Kinase/blood , Creatine Kinase/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Heart Rate/drug effects , Isoproterenol/pharmacology , Lipid Peroxidation/drug effects , Male , Membrane Lipids/metabolism , Myocardial Ischemia/complications , Myocardial Ischemia/pathology , Myocardial Ischemia/prevention & control , Myocardium/enzymology , Myocardium/pathology , Peroxidase/drug effects , Peroxidase/metabolism , Rabbits , Reperfusion Injury/physiopathology , Ventricular Pressure/drug effects
5.
J Cardiovasc Pharmacol ; 36(2): 277-81, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10942172

ABSTRACT

Carvedilol is a vasodilating beta-blocker and antioxidant approved for treatment of mild to moderate hypertension. angina, and congestive heart failure. Metoprolol is a beta1-selective adrenoceptor antagonist. When carvedilol and metoprolol were recently compared in clinical trials for heart failure, each showed beneficial beta-blocker effects such as improved symptoms, quality of life, exercise tolerance, and ejection fraction, with no between-group differences. When thiobarbituric acid reactive substance (TBARS) levels were measured in serum as an indirect marker of free radical activity, there were also no between-group differences. However, we had noted superior cardioprotection by carvedilol in comparison to metoprolol in ischemia and reperfusion models. We therefore examined antioxidant activity directly in cells and tissues. Here we show that in cultured rat cerebellar neurons, and in brain and heart membranes, carvedilol has far greater antioxidant activity than metoprolol, which is essentially inactive as an antioxidant in these model systems. The antioxidant activity of carvedilol could be explained by a greater degree of lipophilicity, as measured by its ClogP value of 3.841 as contrasted to a ClogP value of 1.346 for metoprolol. Alternatively, the molecular structure of carvedilol favors redox recycling, which the structure of metoprolol does not. Therefore, carvedilol could have additional pharmacologic effects that are favorable for long-term therapy.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Antioxidants/pharmacology , Carbazoles/pharmacology , Metoprolol/pharmacology , Propanolamines/pharmacology , Animals , Carvedilol , Cell Membrane/drug effects , Cells, Cultured , Free Radicals/chemistry , Heart/drug effects , Lipid Peroxidation/drug effects , Neurons/drug effects , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Thiobarbituric Acid Reactive Substances/metabolism
6.
Annu Rev Pharmacol Toxicol ; 40: 177-91, 2000.
Article in English | MEDLINE | ID: mdl-10836132

ABSTRACT

Selection and validation of novel molecular targets have become of paramount importance in light of the plethora of new potential therapeutic drug targets that have emerged from human gene sequencing. In response to this revolution within the pharmaceutical industry, the development of high-throughput methods in both biology and chemistry has been necessitated. This review addresses these technological advances as well as several new areas that have been created by necessity to deal with this new paradigm, such as bioinformatics, cheminformatics, and functional genomics. With many of these key components of future drug discovery now in place, it is possible to map out a critical path for this process that will be used into the new millennium.


Subject(s)
Chemistry, Pharmaceutical , Drug Industry , Genetic Therapy , Genome, Human , Animals , Humans
7.
Anal Chem ; 72(8): 1894-904, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10784159

ABSTRACT

We examine the use of thionylphosphazene-based block copolymers as matrixes for oxygen sensor applications. Poly(aminothionylphosphazene)-b-poly(tetrahydrofuran) (PATPy-PTHFx) block copolymers were prepared via reaction of ring-opened poly(chlorothionylphosphazene) with THF and subsequently with excess n-butylamine (to form PBATPy-PTHFx) or methylamine (to form PMATPy-PTHFx). The block copolymers were characterized by NMR, gel permeation chromatography, and differential scanning calorimetry. Films of PBATPy-PTHFx block copolymers containing platinum octaethylporphyrin or [Ru(dpp)3]Cl2 (dpp = 4,7-diphenyl-1,10-phenanthroline) as the oxygen-sensitive chromophore were prepared, and time-scan experiments were carried out to determine the diffusion coefficients, Do2, and solubilities, So2, of oxygen therein. Despite microphase separation, the data fit well to a simple Fick's law description of oxygen diffusion and gave Do2 values smaller than that for the n-butylamino-substituted PBATP635. For films freshly annealed above the melting point of PTHFx, the Do2 values were 35-50% (dye-dependent) larger than after aging 3 days at room temperature. Films with [Ru(dpp)3]Cl2 as the dye were evaluated as media for phosphorescent pressure-sensing. The dye-containing polymer films exhibit linear Stern-Volmer-like plots, even at high dye concentrations, as well as good photostability, and significantly higher sensitivity to oxygen quenching than simple mixtures of the analogous homopolymers.

8.
Am Heart J ; 138(3 Pt 2): 246-51, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10467220

ABSTRACT

Selective blockade of the angiotensin II AT1 receptor represents a novel mechanism for interrupting the renin-angiotensin system without altering the potential benefits of AT2 receptor stimulation. This selective inhibition produces none of the disadvantages associated with reduced bradykinin metabolism and angiotensin II generated by non-angiotensin-converting enzyme pathways. Eprosartan is a potent (1.4 nmol/L) AT1 receptor antagonist that competitively blocks angiotensin II-induced vascular contraction. In various animal models of disease, including hypertension and stroke, eprosartan is effective in reducing disease progression. Eprosartan also has sympathoinhibitory activity, as demonstrated by an inhibition of the pressor responses induced by activation of sympathetic outflow through spinal cord stimulation in pithed rats. In contrast, some of the other angiotensin II receptor antagonists, such as losartan, at equivalent angiotensin II blocking doses, have no effect on sympathetic nervous system activity. Because eprosartan can inhibit both the direct effects of angiotensin II as well as the indirect effects that are mediated by enhanced sympathetic neurotransmission, this may represent an important advance in the treatment of elevated systolic blood pressure.


Subject(s)
Acrylates/pharmacology , Antihypertensive Agents/pharmacology , Hypertension/drug therapy , Imidazoles/pharmacology , Receptors, Angiotensin/drug effects , Renin-Angiotensin System/physiology , Thiophenes , Angiotensin II/pharmacology , Angiotensin Receptor Antagonists , Animals , Blood Pressure/physiology , Cerebrovascular Disorders/prevention & control , Disease Models, Animal , Humans , Rats , Receptors, Angiotensin/physiology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiology
9.
J Hypertens Suppl ; 17(2): S27-32, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10465064

ABSTRACT

Systolic hypertension is a major risk factor for cardiovascular disease. The determinants of systolic blood pressure are peripheral resistance and arterial compliance. Arterial vasoconstriction, vascular growth and fluid retention, induced by the renin-angiotensin system directly or indirectly by enhancing sympathetic nervous system activity, are important factors in increasing peripheral resistance, decreasing arterial compliance and, consequently, elevating systolic blood pressure. Selective blockade of the angiotensin II type 1 (AT1) receptor represents a novel mechanism for interrupting the renin-angiotensin system. This provides the additional benefit of blocking angiotensin II generated by non-angiotensin-converting-enzyme pathways without altering either bradykinin metabolism or the potential beneficial effects of AT2 receptor stimulation. Eprosartan is a potent (1.4 nmol/l) AT1 receptor antagonist that inhibits angiotensin-II-induced vascular contraction in a competitive manner. Eprosartan is effective in reducing disease progression in animal models of hypertension, heart failure, renal disease and stroke. Furthermore, eprosartan causes a large increase in arterial compliance in hypertensive rats fed high-salt and high-fat diets. Eprosartan also possesses sympathoinhibitory activity as demonstrated by an inhibition of the pressor responses induced by activation of sympathetic outflow through spinal cord stimulation in pithed rats. In contrast, other angiotensin II receptor antagonists, such as losartan, used at equivalent angiotensin II blocking activity, do not appear to alter sympathetic nervous system activity. Angiotensin II receptor antagonists, such as eprosartan, that have the ability to block both the direct effects of angiotensin II and the indirect effects mediated by enhanced sympathetic neurotransmission, may represent an important advance in the treatment of elevated systolic blood pressure.


Subject(s)
Acrylates/pharmacology , Angiotensin Receptor Antagonists , Antihypertensive Agents/pharmacology , Hypertension/drug therapy , Imidazoles/pharmacology , Thiophenes , Animals , Blood Pressure/drug effects , Humans , Hypertension/metabolism , Hypertension/physiopathology , Renin-Angiotensin System/drug effects , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiopathology , Systole , Treatment Outcome , Vascular Resistance/drug effects
10.
Curr Opin Chem Biol ; 3(4): 474-80, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10419841

ABSTRACT

The realization that apoptosis is genetically programmed raises the exciting prospect that modulating apoptosis may provide novel approaches for treatment of cardiovascular diseases in which apoptosis has been demonstrated. Low molecular weight inhibitors of caspases and mitogen-activated protein kinases have been evaluated, with promising results in a variety of cardiovascular apoptotic models.


Subject(s)
Apoptosis , Cardiovascular Diseases/drug therapy , Animals , Apoptosis/drug effects , Cardiovascular Diseases/pathology , Enzyme Inhibitors/pharmacology , Humans , Signal Transduction
11.
Eur Urol ; 36 Suppl 1: 17-22, 1999.
Article in English | MEDLINE | ID: mdl-10393468

ABSTRACT

Although the selective alpha1-adrenoceptor antagonists were initially developed as antihypertensive drugs, and they are still utilized for this indication, the alpha1-adrenoceptor blockers are now used extensively for the symptomatic treatment of benign prostatic hyperplasia (BPH). As a result, a number of new drugs in this class have been specifically developed for use in BPH. The utility of alpha1-adrenoceptor antagonists in BPH derives from the observation, made several decades ago, that the irreversible, alpha1- adrenoceptor selective antagonist phenoxybenzamine, blocked the contractile activity of norepinephrine in isolated strips of rat or human prostate. Following the further subclassification of alpha1-adrenoceptors into the alpha1A-, alpha1B- and alpha1D-adrenoceptor subtypes, the relationship between subtype selectivity and efficacy in BPH has been investigated in the hope of developing more selective drugs for the treatment of this disorder. Molecular characterization of the adrenoceptor population in human prostate clearly shows the alpha1A-adrenoceptor subtype to predominate, and highly selective alpha1A-adrenoceptor antagonists have been identified and investigated in BPH. However, controversy remains as to whether prostatic smooth muscle contraction is mediated by the alpha1A-adrenoceptor, or by another novel alpha1-adrenoceptor subtype (not corresponding to any of the three known recombinant alpha1-adrenoceptors), or both. alpha1-Adrenoceptor agonists have been used clinically for the treatment of stress incontinence, acting to increase urethral tone by contracting urethral smooth muscle. Research efforts are ongoing to identify agents of this class having a selective action on urethral versus vascular smooth muscle, in order to produce a greater effect on the urethra without producing dose-limiting increases in blood pressure. Local administration of vascular smooth muscle relaxants, either alone or in combination, has been used for the treatment of erectile dysfunction. An alpha1-adrenoceptor antagonist is often used as one comportent in such mixtures, which act to relax trabecular smooth muscle. The recent demonstration that a systemically administered drug can produce a sufficiently selective action on cavernosal smooth muscle to allow efficacy without producing limiting systemic side effects has renewed interest in the possibility of systemic administration of alpha1-adrenoceptor antagonists for this indication.


Subject(s)
Prostatic Hyperplasia/drug therapy , Receptors, Adrenergic, alpha-1/physiology , Urogenital System/physiology , Adrenergic alpha-Agonists/therapeutic use , Adrenergic alpha-Antagonists/therapeutic use , Animals , Erectile Dysfunction/drug therapy , Humans , Male , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Prostatic Hyperplasia/physiopathology , Urinary Incontinence/drug therapy , Urogenital System/drug effects
12.
Eur J Pharmacol ; 373(1): 51-62, 1999 May 28.
Article in English | MEDLINE | ID: mdl-10408251

ABSTRACT

We evaluated the uroselectivity of a series of alpha1-adrenoceptor antagonists by comparing their potency against phenylephrine-induced increases in urethral perfusion pressure and diastolic blood pressure in the anesthetized rabbit and pithed rat. In the rabbit, Rec 15/2739 (N-[3-[4-(2-methoxyphenyl)-1-piperazinyl]propyl]-3-methyl-4-oxo-2-phenyl -4H-1-benzopyran-8-carboxamide) as well as analogs with a chlorine substituent on the methoxyphenyl ring (Rec 15/2869) or this substituent combined with the replacement of the phenyl substituent on the pyran ring by cyclohexyl (Rec 15/3011) were 2-6-fold more potent against the urethral vs. vascular response to phenylephrine. Rec 15/2841 (N-[3-[4-(2-methoxyphenyl)-1-piperazinyl]propyl]-3-methyl-4-oxo-2-cyc lohexy-4H-1-benzopyran-8-carboxamide) was only 1.5-fold more potent against the urethral response. SL 89.0591 and tamsulosin also showed selectivity for the urethral response (2-2.5-fold), while the quinazolines produced equipotent blockade of urethral and vascular responses (selectivity ratio = 0.9-1.1). The urethral selectivities of Rec 15/2739 and its derivatives were confirmed by evaluation of the response to tilt in sedated, hypovolemic rabbits. Phenylephrine challenge assays did not show any of the antagonists, with the exception of terazosin at 300 microg kg(-1), to be uroselective in the rat (selectivity ratios = 0.2-1.5); potentiation of tilt-induced hypotension in the anesthetized rat showed substantial differences from the rabbit, with Rec 15/2739, but not Rec 15/3011 and Rec 15/2841 showing orthostatic effects equivalent to that observed for prazosin. Hence, Rec 15/2739 was uroselective in the rabbit, but not in the rat, while two of its close structural analogs were highly uroselective in both species. An assay for orthostatic activity in the conscious rat yielded different results, showing prazosin and terazosin, but not Rec 15/2739, to cause a reversal of the pressor response to tilt. Hence, the apparent uroselectivity of an alpha1-adrenoceptor antagonist is both species- and assay-dependent.


Subject(s)
Adrenergic Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Blood Pressure/drug effects , Posture/physiology , Receptors, Adrenergic, alpha-1/drug effects , Anesthesia , Animals , Blood Pressure/physiology , Chromones/chemistry , Chromones/pharmacology , Consciousness , Decerebrate State , Diastole , Dose-Response Relationship, Drug , Hypnotics and Sedatives/pharmacology , Hypotension, Orthostatic/physiopathology , Male , Phenylephrine/pharmacology , Quinazolines/pharmacology , Rabbits , Rats , Species Specificity , Structure-Activity Relationship , Urethra/drug effects , Urethra/physiology
13.
J Pharmacol Exp Ther ; 289(1): 48-53, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10086986

ABSTRACT

Clinical studies conducted with carvedilol suggest that beta-adrenoceptor antagonism is an effective therapeutic approach to the treatment of heart failure. However, many beta-adrenoceptor antagonists are weak partial agonists and possess significant intrinsic sympathomimetic activity (ISA), which may be problematic in the treatment of heart failure. In the present study, the ISAs of bucindolol, xamoterol, bisoprolol, and carvedilol were evaluated and compared in normal rats [Sprague-Dawley (SD)], in rats with confirmed heart failure [spontaneously hypertensive heart failure (SHHF)], and in isolated neonatal rat cardiomyocytes. At equieffective beta1-adrenolytic doses, the administration of xamoterol and bucindolol produced a prolonged, equieffective, and dose-related increase in heart rate in both pithed SD rats (ED50 = 5 and 40 microgram/kg, respectively) and SHHF rats (ED50 = 6 and 30 microgram/kg, respectively). The maximum effect of both compounds in SHHF rats was approximately 50% of that observed in SD rats. In contrast, carvedilol and bisoprolol had no significant effect on resting heart rate in the pithed SD or SHHF rat. The maximum increase in heart rate elicited by xamoterol and bucindolol was inhibited by treatment with propranolol, carvedilol, and betaxolol (beta1-adrenoceptor antagonist) but not by ICI 118551 (beta2-adrenoceptor antagonist) in neonatal rat. When the beta-adrenoceptor-mediated cAMP response was examined in cardiomyocytes, an identical partial agonist/antagonist response profile was observed for all compounds, demonstrating a strong correlation with the in vivo results. In contrast, GTP-sensitive ligand binding and tissue adenylate cyclase activity were not sensitive methods for detecting beta-adrenoceptor partial agonist activity in the heart. In summary, xamoterol and bucindolol, but not carvedilol and bisoprolol, exhibited direct beta1-adrenoceptor-mediated ISA in normal and heart failure rats.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Heart Failure/physiopathology , Heart Rate/drug effects , Adrenergic beta-Agonists/pharmacology , Animals , Animals, Newborn , Bisoprolol/pharmacology , Carbazoles/pharmacology , Carvedilol , Cell Membrane/metabolism , Cells, Cultured , Cyclic AMP/metabolism , Decerebrate State , Dose-Response Relationship, Drug , Heart Failure/metabolism , Iodocyanopindolol , Ligands , Male , Myocardium/metabolism , Propanolamines/pharmacology , Radioligand Assay , Rats , Rats, Sprague-Dawley , Xamoterol/pharmacology
14.
Proc West Pharmacol Soc ; 41: 225-8, 1998.
Article in English | MEDLINE | ID: mdl-9836297

ABSTRACT

The catecholamines were found to inhibit the binding of the alpha 2-adrenoceptor agonist, [3H]-clonidine, to the recombinant wild type alpha 2a-adrenoceptor (Table 1) with potencies that are consistent with their functional activity in alpha 2-adrenoceptor test systems [6,7]. Mutation of Ser165 to alanine had no significant effect (less than 2-fold) on the affinity of any of the catecholamines for the alpha 2a-adrenoceptor, and in particular, the ratios of affinities between the corresponding (-)- and (+)-enantiomers of the catecholamines were not altered by the point mutation at Ser165. These findings indicate clearly that Ser165, in contrast to predictions made by molecular modeling, plays little if any role in the binding of the catecholamines in general, and cannot be involved in the attachment of the beta-hydroxyl group to the alpha 2a-adrenoceptor. Mutation of either Ser90 on transmembrane helix II or Ser419 on transmembrane helix VII to alanine produced dramatic and selective reductions in the affinity of the (-)-enantiomers of the catecholamines for the alpha 2a-adrenoceptor, with no changes occurring in affinities of the (+)-enantiomers. Thus, the affinities of (-)-norepinephrine and (-)-epinephrine for the Ser90 and Ser419 mutants of the alpha 2a-adrenoceptor were 35-75 fold lower than their affinities for the wild type receptor (Table 1), suggesting that Ser90 and/or Ser419 are involved in the attachment of the beta-hydroxyl groups of the catecholamines to the receptor. Similarly, the affinity of (+/-)-6-fluoronorepinephrine was reduced by 100-fold for the Ser90 mutant receptor (Table 1). Importantly, the affinities of the (+)-enantiomers of the catecholamines, as well as dopamine and epinine, which are the corresponding analogs of norepinephrine and epinephrine which lack the beta-hydroxyl group, were not affected by mutation of Ser90 or Ser419 to alanine (Table 1). Asn293 in transmembrane helix VI has also been proposed to be involved in the interaction of the beta-hydroxyl group of isoproterenol with the beta 2-adrenoceptor [4]. The alpha 2a-adrenoceptor contains three hydroxyl bearing amino acids at a position corresponding to this site (Thr393-Tyr394-Thr395). These amino acids could theoretically form a hydrogen bond with the beta-hydroxyl group of a catecholamine, and therefore could serve as a potential point of attachment. Simultaneous mutation of all three of these amino acids to Ala-Phe-Ala reduced the affinity of the (-)-enantiomers of the catecholamines by 12-20 fold, which is somewhat less than what was observed for mutation of either Ser90 or Ser419 (Table 1). However, in contrast to mutation of Ser90 or Ser419, which had no effect on the affinity of the (+)-enantiomers, mutation of the three residues in transmembrane helix VI did significantly reduce the affinities of the (+)-enantiomers of the catecholamines by approximately 5- to 9-fold, indicating that mutations at these points of the receptor are not selective for the (-)-enantiomers, and are therefore not likely to be involved in the attachment of the beta-hydroxyl group of the catecholamines.


Subject(s)
Catecholamines/metabolism , Cell Membrane/physiology , Receptors, Adrenergic/metabolism , Serine/metabolism , Molecular Conformation
15.
Eur J Pharmacol ; 356(2-3): 193-8, 1998 Sep 04.
Article in English | MEDLINE | ID: mdl-9774249

ABSTRACT

The aim of this study was to investigate the effect of SB 211475, a metabolite of carvedilol with weak alpha1-adrenoceptor antagonism and antioxidant effect, on myocardial reperfusion injury and infarct size in anesthetized rabbits. The rabbits were subjected to 60 min of regional myocardial ischemia and 180 min of reperfusion. SB 211475 was administered either as 0.3, 1.0 or 3.0 mg/kg and compared to vehicle and carvedilol (1 mg/kg) treated animals. The lowest dose of SB 211475 (0.3 mg/kg) did not reduce infarct size compared to vehicle, whereas SB 211475 1.0 or 3.0 mg/kg reduced infarct size significantly compared to vehicle (41.2 +/- 2.2% and 40.5 +/- 2.8% vs. 59.1 +/- 3.9%, p < 0.05). Carvedilol reduced infarct size significantly more than SB 211475 1.0 and 3.0 mg/kg (28.8 +/- 3.9% vs. 41.2 +/- 2.2% and 40.5 +/- 2.7%, p < 0.05). Carvedilol and SB 211475 1.0 and 3.0 mg/kg reduced myeloperoxidase activity to the same extent, indicative of reduced inflammation. Rate-pressure product did not differ between doses of SB 211475. In conclusion, SB 211475 in the two highest doses reduced infarct size by protecting from reperfusion injury, possibly by reduced neutrophil accumulation. The superior cardiac protective effect of carvedilol over SB 211475 are most likely due to its adrenergic pharmacology including non-selective beta- and alpha1-adrenoceptor antagonism.


Subject(s)
Adrenergic alpha-Antagonists/therapeutic use , Antioxidants/therapeutic use , Carbazoles/therapeutic use , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/drug therapy , Propanolamines/therapeutic use , Animals , Antioxidants/administration & dosage , Carbazoles/administration & dosage , Carvedilol , Dose-Response Relationship, Drug , Hemodynamics/drug effects , Male , Myocardial Reperfusion Injury/prevention & control , Peroxidase/metabolism , Propanolamines/administration & dosage , Rabbits
16.
Hypertension ; 32(4): 746-52, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9774374

ABSTRACT

Activation of the renin-angiotensin system may contribute to the derangement in renal and cardiac function in congestive heart failure. The present study evaluated the effects of eprosartan, a selective angiotensin II receptor antagonist, on renal hemodynamic and excretory parameters and on the development of cardiac hypertrophy in rats with aortocaval fistula, an experimental model of congestive heart failure. Infusion of eprosartan (1.0 mg/kg) in rats with aortocaval fistula produced a significant increase (+34%) in total renal blood flow and a sustained decrease (-33%) in the calculated renal vascular resistance. These effects on renal hemodynamics were more pronounced than those observed in sham-operated control rats and occurred despite a significant fall (-12%) in mean arterial blood pressure. Moreover, eprosartan caused a preferential increase in renal cortical blood perfusion and significantly increased glomerular filtration in rats with congestive heart failure. Chronic administration of eprosartan (5.0 mg/kg per day for 7 days through osmotic minipumps inserted intraperitoneally on the day of operation) resulted in a significant enhancement of urinary sodium excretion compared with nontreated rats with heart failure. Moreover, administration of eprosartan to salt-retaining rats with congestive heart failure resulted in a progressive increase and ultimate recovery in urinary sodium excretion. Finally, early treatment with eprosartan blocked the development of cardiac hypertrophy in rats with aortocaval fistula to a larger extent than the angiotensin-converting enzyme inhibitor enalapril. These findings emphasize the importance of angiotensin II in mediating the impairment in renal function and induction of cardiac hypertrophy in heart failure and further suggest that angiotensin II receptor blockade may be a useful treatment of these consequences in severe cardiac failure.


Subject(s)
Acrylates/therapeutic use , Angiotensin Receptor Antagonists , Antihypertensive Agents/therapeutic use , Cardiomegaly/prevention & control , Heart Failure/drug therapy , Hemodynamics/drug effects , Imidazoles/therapeutic use , Kidney/drug effects , Thiophenes , Acrylates/pharmacology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Antihypertensive Agents/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Enalapril/therapeutic use , Heart Failure/physiopathology , Imidazoles/pharmacology , Male , Rats , Rats, Wistar , Renal Circulation/drug effects , Sodium/urine
17.
J Cardiovasc Pharmacol ; 32 Suppl 1: S22-30, 1998.
Article in English | MEDLINE | ID: mdl-9731692

ABSTRACT

A variety of pathophysiologic processes are activated in patients with congestive heart failure (CHF), and some of these have been implicated in the progression of the disease. The most important processes to be activated in CHF are the neurohormonal systems, which include the renin-angiotensin system, the sympathetic nervous system, and the endothelin system. In addition to the neurohormonal systems, the formation of reactive oxygen free radicals is increased in patients with CHF. It has been postulated that stimulation of neurohormonal pathways and the formation of oxygen free radicals ultimately lead to the activation of a family of transcription factors that are involved in cardiac remodeling, which is a hallmark of CHF. In addition, the formation of oxygen free radicals has been implicated in the process of apoptosis or programmed cell death, which may be responsible for a continued loss of myocardial cells, resulting in the progressive decrease in left ventricular function that occurs over time in patients with CHF. Carvedilol is a multiple-action neurohormonal antagonist that is effective in slowing the progression of CHF. In double-blind, placebo-controlled clinical trials, carvedilol decreased mortality by 65% (p <0.001) and significantly reduced hospitalization. Carvedilol is a nonselective beta-blocker and vasodilator, the latter activity resulting from alpha1-adrenoceptor blockade. The hemodynamic responses produced by carvedilol result primarily from the blockade of beta1-, beta2-, and alpha1-adrenoceptors. Carvedilol reduces total peripheral vascular resistance and preload without significantly compromising cardiac output or eliciting reflex tachycardia. Carvedilol is also a potent antioxidant that may protect the myocardium from damage produced by oxygen radicals and, as a consequence of its antioxidant activity, carvedilol also inhibits apoptosis in the myocardium. The ability of carvedilol to inhibit apoptosis in the heart may be responsible, in part, for the ability of the drug to reduce mortality and to inhibit the progression of CHF.


Subject(s)
Apoptosis , Heart Failure/etiology , Reactive Oxygen Species/metabolism , Adrenergic beta-Antagonists/therapeutic use , Antioxidants/pharmacology , Carbazoles/therapeutic use , Carvedilol , Disease Progression , Heart Failure/drug therapy , Heart Failure/metabolism , Heart Failure/pathology , Hemodynamics/drug effects , Humans , Propanolamines/therapeutic use , Receptors, Adrenergic, beta/drug effects
18.
J Pharmacol Exp Ther ; 286(3): 1191-6, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9732378

ABSTRACT

Administration of the selective adenosine A1 receptor antagonist, CVT-124, to conscious chronically instrumented rats resulted in significant increases in urine flow rate and sodium excretion without affecting potassium excretion or renal hemodynamics. Its maximum effect was twice that of hydrochlorothiazide which was associated with a significant kaliuresis. The diuretic effect of CVT-124 was less than that observed with furosemide; however, furosemide administration was associated with a large increase in potassium excretion as well as a reduction in glomerular filtration rate. When given at equinatriuretic doses, CVT-124 enhanced the diuretic and natriuretic activity of furosemide without further increasing potassium excretion. In contrast, the combination of hydrochlorothiazide and furosemide resulted in a 3-fold increase in potassium excretion. These data suggest that CVT-124 possesses unique diuretic activity and, as such, it represents a potential new therapeutic in fluid retaining disorders. In addition, its unique mechanism of action suggests that CVT-124 would be effective in otherwise diuretic-resistant patients.


Subject(s)
Diuretics/pharmacology , Purinergic P1 Receptor Antagonists , Xanthines/pharmacology , Animals , Dose-Response Relationship, Drug , Furosemide/pharmacology , Glomerular Filtration Rate/drug effects , Hydrochlorothiazide/pharmacology , Male , Rats , Rats, Sprague-Dawley , Renal Circulation/drug effects
19.
Prog Cardiovasc Dis ; 41(1 Suppl 1): 17-24, 1998.
Article in English | MEDLINE | ID: mdl-9715819

ABSTRACT

Carvedilol is a novel cardiovascular drug of proven efficacy in the treatment of hypertension, angina, and heart failure. Several mechanisms may account for the beneficial effects of carvedilol in patients with heart failure. As with other beta-blockers, blockade of cardiac beta-adrenergic receptors (both beta1 and beta2), and hence reduction of cardiac work load and oxygen consumption, plays an important role in the actions of this agent. Additional benefit is provided by vasodilation (alphal-adrenergic blockage) at peripheral resistance vessels, which decreases preload and after-load, thereby further reducing cardiac work and wall tensions. In addition, potential advantages of carvedilol resulting from alpha1-adrenergic blockade are likely because alpha1-adrenergic receptors mediate cardiac remodeling by inducing hypertrophy. Finally, carvedilol is a potent antioxidant and is unique among beta-blockers in this respect. In recent years, evidence has accumulated in support of the role played by reactive oxygen radicals in chronic pathological states of the myocardium. In this article, the role of oxygen radicals in heart failure is discussed with special reference to apoptosis, a phenomenon believed to be involved in progressive cardiac myocyte loss in ischemic or myopathic heart diseases. The potential role of the antioxidant actions of carvedilol, especially in prevention of apoptotic cell death, is highlighted as a novel mechanism of action in heart failure.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Apoptosis/drug effects , Carbazoles/therapeutic use , Free Radical Scavengers/therapeutic use , Heart Failure/drug therapy , Propanolamines/therapeutic use , Reactive Oxygen Species , Animals , Antioxidants/therapeutic use , Carvedilol , Free Radicals , Heart Failure/metabolism , Heart Failure/pathology , Humans , Myocardium/metabolism , Oxidative Stress
20.
Eur J Pharmacol ; 351(3): 341-50, 1998 Jun 26.
Article in English | MEDLINE | ID: mdl-9721026

ABSTRACT

Carvedilol, a selective alpha1 and non-selective beta-adrenoceptor antagonist and antioxidant, has been shown to provide significant cardiac protection in animal models of myocardial ischemia. To further explore the mechanisms contributing to carvedilol cardioprotection efficacy, the effects of carvedilol on hemodynamic variables, infarct size and myeloperoxidase activity (an index of neutrophil accumulation) were compared with a beta1-selective adrenoceptor antagonist, metoprolol. Carvedilol (1 mg/kg) or metoprolol (1 mg/kg or 1 mg/kg + 0.5 mg/kg 90 min later) was given intravenously 5 min before reperfusion. In vehicle-treated rabbits, ischemia (60 min) and reperfusion (180 min) resulted in significant increments in left ventricular end diastolic pressure, large infarcts (59+/-2.6% of area-at-risk) and marked increase in myeloperoxidase activity (0.59+/-0.09 U/100 mg tissue). Carvedilol treatment resulted in sustained reduction of pressure-rate-index and significantly smaller infarcts (22.0+/-2.5%, P < 0.01 vs. vehicle) as well as decreased myeloperoxidase activity (0.186+/-0.056 U/100 mg tissue, P < 0.01 vs. vehicle). The highest dose of metoprolol, 1 mg/kg + 0.5 mg/kg, that resulted in pressure-rate-index comparable to that of 1.0 mg/kg carvedilol, failed to reduce myeloperoxidase activity in the ischemic myocardial tissue, and the infarct size (35+/-3.1%) was significantly larger than in carvedilol-treated animals. Taken together, this study suggests that the superior cardioprotection of carvedilol over metoprolol is not a consequence of hemodynamic variances but possibly the result of the additional pharmacological properties of carvedilol such as the antioxidant and anti-neutrophil effects.


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Antioxidants/pharmacology , Carbazoles/pharmacology , Metoprolol/pharmacology , Myocardial Ischemia/prevention & control , Myocardial Reperfusion Injury/prevention & control , Propanolamines/pharmacology , Adrenergic alpha-Antagonists/chemistry , Adrenergic alpha-Antagonists/metabolism , Adrenergic beta-Antagonists/chemistry , Adrenergic beta-Antagonists/metabolism , Animals , Antioxidants/chemistry , Antioxidants/metabolism , Blood Pressure/drug effects , Carbazoles/chemistry , Carbazoles/metabolism , Carvedilol , Heart Rate/drug effects , Male , Metoprolol/chemistry , Metoprolol/metabolism , Myocardial Infarction/etiology , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Myocardial Ischemia/complications , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/enzymology , Myocardium/pathology , Oxidation-Reduction , Peroxidase/metabolism , Propanolamines/chemistry , Propanolamines/metabolism , Propranolol/chemistry , Propranolol/metabolism , Propranolol/pharmacology , Rabbits , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Recombinant Proteins/metabolism , Solubility , Ventricular Function, Left/drug effects , Ventricular Pressure/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...