Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Peripher Nerv Syst ; 19(2): 77-87, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24934510

ABSTRACT

NIDDK, JDRF, and the Diabetic Neuropathy Study Group of EASD sponsored a meeting to explore the current status of animal models of diabetic peripheral neuropathy. The goal of the workshop was to develop a set of consensus criteria for the phenotyping of rodent models of diabetic neuropathy. The discussion was divided into five areas: (1) status of commonly used rodent models of diabetes, (2) nerve structure, (3) electrophysiological assessments of nerve function, (4) behavioral assessments of nerve function, and (5) the role of biomarkers in disease phenotyping. Participants discussed the current understanding of each area, gold standards (if applicable) for assessments of function, improvements of existing techniques, and utility of known and exploratory biomarkers. The research opportunities in each area were outlined, providing a possible roadmap for future studies. The meeting concluded with a discussion on the merits and limitations of a unified approach to phenotyping rodent models of diabetic neuropathy and a consensus formed on the definition of the minimum criteria required for establishing the presence of the disease. A neuropathy phenotype in rodents was defined as the presence of statistically different values between diabetic and control animals in 2 of 3 assessments (nocifensive behavior, nerve conduction velocities, or nerve structure). The participants propose that this framework would allow different research groups to compare and share data, with an emphasis on data targeted toward the therapeutic efficacy of drug interventions.


Subject(s)
Consensus , Diabetic Neuropathies/physiopathology , Phenotype , Animals , Behavior, Animal/physiology , Biomedical Research/methods , Biomedical Research/standards , Diabetic Neuropathies/pathology , Disease Models, Animal , Humans , Neural Conduction/physiology , Peripheral Nerves/pathology
2.
Diabetologia ; 54(5): 1242-51, 2011 May.
Article in English | MEDLINE | ID: mdl-21267539

ABSTRACT

AIMS/HYPOTHESIS: The aim of the study was to investigate the effects of genetic deficiency of aldose reductase in mice on the development of key endpoints of diabetic nephropathy. METHODS: A line of Ar (also known as Akr1b3)-knockout (KO) mice, a line of Ar-bitransgenic mice and control C57BL/6 mice were used in the study. The KO and bitransgenic mice were deficient for Ar in the renal glomeruli and all other tissues, with the exception of, in the bitransgenic mice, a human AR cDNA knockin-transgene that directed collecting-tubule epithelial-cell-specific AR expression. Diabetes was induced in 8-week-old male mice with streptozotocin. Mice were further maintained for 17 weeks then killed. A number of serum and urinary variables were determined for these 25-week-old mice. Periodic acid-Schiff staining, western blots, immunohistochemistry and protein kinase C (PKC) activity assays were performed for histological analyses, and to determine the levels of collagen IV and TGF-ß1 and PKC activities in renal cortical tissues. RESULTS: Diabetes-induced extracellular matrix accumulation and collagen IV overproduction were completely prevented in diabetic Ar-KO and bitransgenic mice. Ar deficiency also completely or partially prevented diabetes-induced activation of renal cortical PKC, TGF-ß1 and glomerular hypertrophy. Loss of Ar results in a 43% reduction in urine albumin excretion in the diabetic Ar-KO mice and a 48% reduction in the diabetic bitransgenic mice (p < 0.01). CONCLUSIONS/INTERPRETATION: Genetic deficiency of Ar significantly ameliorated development of key endpoints linked with early diabetic nephropathy in vivo. Robust and specific inhibition of aldose reductase might be an effective strategy for the prevention and treatment of diabetic nephropathy.


Subject(s)
Aldehyde Reductase/deficiency , Aldehyde Reductase/genetics , Diabetic Nephropathies/genetics , Animals , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetic Nephropathies/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
3.
J Med Chem ; 44(17): 2695-700, 2001 Aug 16.
Article in English | MEDLINE | ID: mdl-11495581

ABSTRACT

We report here on our medicinal chemistry and pharmacology efforts to provide a potent sorbitol dehydrogenase inhibitor (SDI) as a tool to probe a recently disclosed hypothesis centered on the role of sorbitol dehydrogenase (SDH) in the second step of the polyol pathway, under conditions of high glucose flux. Starting from a weak literature lead, 2, and through newly developed structure-activity relationships, we have designed and executed an unambiguous synthesis of enantiomeric SDI, 6, which is at least 10x more potent than 2. Also, 6 potently inhibits SDH in streptozotocin-diabetic rat sciatic nerve. We have described an expedient synthesis of a key building template, 33, for future research in the SDI area that may facilitate the discovery of even more potent SDIs with longer duration of action in vivo.


Subject(s)
Enzyme Inhibitors/chemical synthesis , L-Iditol 2-Dehydrogenase/antagonists & inhibitors , Pyrimidines/chemical synthesis , Sulfonamides/chemical synthesis , Administration, Oral , Animals , Diabetes Mellitus, Experimental/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Fructose/antagonists & inhibitors , Fructose/biosynthesis , Humans , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Rats , Sciatic Nerve/metabolism , Stereoisomerism , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Tissue Distribution
4.
Am J Physiol Heart Circ Physiol ; 279(4): H1447-52, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11009428

ABSTRACT

This study investigated whether aldose reductase (AR) inhibition with zopolrestat, either alone or in combination with an adenosine A(3)-receptor agonist (CB-MECA), reduced myocardial ischemic injury in rabbit hearts subjected to 30 min of regional ischemia and 120 min of reperfusion. Zopolrestat reduced infarct size by up to 61%, both in vitro (2 nM to 1 microM; EC(50) = 24 nM) and in vivo (50 mg/kg). Zopolrestat reduced myocardial sorbitol concentration (index of AR activity) by >50% (control, 15.0 +/- 2.2 nmol/g; 200 nM zopolrestat, 6.7 +/- 1.3 nmol/g). A modestly cardioprotective concentration of CB-MECA (0.2 nM) allowed a 50-fold reduction in zopolrestat concentration while providing a similar reduction in infarct size (infarct area/area at risk: control, 62 +/- 2%; 1 microM zopolrestat, 24 +/- 5%; 20 nM zopolrestat plus 0.2 nM CB-MECA, 20 +/- 4%). In conclusion, AR inhibition is cardioprotective both in vitro and in vivo. Furthermore, combining zopolrestat with an A(3) agonist allows a reduction in the zopolrestat concentration while maintaining an equivalent degree of cardioprotection.


Subject(s)
Adenosine/analogs & derivatives , Adenosine/pharmacology , Aldehyde Reductase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Myocardial Ischemia/pathology , Phthalazines/pharmacology , Purinergic P1 Receptor Agonists , Thiazoles/pharmacology , Animals , Benzothiazoles , Dose-Response Relationship, Drug , In Vitro Techniques , Male , Myocardium/metabolism , Myocardium/pathology , Rabbits , Receptor, Adenosine A3 , Sorbitol/metabolism
6.
Nature ; 404(6779): 787-90, 2000 Apr 13.
Article in English | MEDLINE | ID: mdl-10783895

ABSTRACT

Diabetic hyperglycaemia causes a variety of pathological changes in small vessels, arteries and peripheral nerves. Vascular endothelial cells are an important target of hyperglycaemic damage, but the mechanisms underlying this damage are not fully understood. Three seemingly independent biochemical pathways are involved in the pathogenesis: glucose-induced activation of protein kinase C isoforms; increased formation of glucose-derived advanced glycation end-products; and increased glucose flux through the aldose reductase pathway. The relevance of each of these pathways is supported by animal studies in which pathway-specific inhibitors prevent various hyperglycaemia-induced abnormalities. Hyperglycaemia increases the production of reactive oxygen species inside cultured bovine aortic endothelial cells. Here we show that this increase in reactive oxygen species is prevented by an inhibitor of electron transport chain complex II, by an uncoupler of oxidative phosphorylation, by uncoupling protein-1 and by manganese superoxide dismutase. Normalizing levels of mitochondrial reactive oxygen species with each of these agents prevents glucose-induced activation of protein kinase C, formation of advanced glycation end-products, sorbitol accumulation and NFkappaB activation.


Subject(s)
Hyperglycemia/etiology , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Animals , Aspartic Acid/metabolism , Blood Glucose/metabolism , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Carrier Proteins/pharmacology , Cattle , Electron Transport , Electron Transport Complex II , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Enzyme Activation , Glycation End Products, Advanced/metabolism , Hyperglycemia/metabolism , Hyperglycemia/pathology , Ion Channels , Malates/metabolism , Membrane Proteins/pharmacology , Mitochondrial Proteins , Multienzyme Complexes/metabolism , NF-kappa B/metabolism , Oxidoreductases/metabolism , Protein Kinase C/metabolism , Rotenone/pharmacology , Sorbitol/metabolism , Succinate Dehydrogenase/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase/pharmacology , Thenoyltrifluoroacetone/analogs & derivatives , Thenoyltrifluoroacetone/pharmacology , Uncoupling Agents/pharmacology , Uncoupling Protein 1
7.
Cardiovasc Res ; 42(1): 130-9, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10435004

ABSTRACT

OBJECTIVE: We have previously demonstrated that zopolrestat, an inhibitor of the enzyme aldose reductase, reduces ischemic injury in hearts from diabetic and non-diabetic rats. To further explore potential cardioprotective mechanisms of zopolrestat, we measured changes in intracellular sodium, calcium, and Na+,K(+)-ATPase activity in zopolrestat treated hearts during ischemia and reperfusion. METHODS: Hearts from acute diabetic (Type I) and age-matched control rats were isolated and retrogradely perfused. Hearts had either control perfusion or exposure to 1 microM zopolrestat for 10 min, followed by 20 min of global ischemia and 60 min of reperfusion. Changes in intracellular sodium and calcium were measured using 23Na and 19F magnetic resonance spectroscopy, respectively, while the activity of Na+,K(+)-ATPase was measured using biochemical assays. RESULTS: Zopolrestat blunted the rise in [Na]i during ischemia in both diabetic hearts and non-diabetic hearts. The end-ischemic [Na]i was 21.3 +/- 2.6 mM in the zopolrestat treated diabetics and 25.9 +/- 2.3 in zopolrestat treated non-diabetics, versus 31.6 +/- 2.6 mM and 32.9 +/- 2.8 mM in the untreated diabetics and untreated non-diabetics, respectively, (P = 0.002). Similarly, the rise in [Ca]i at the end of ischemia was significantly reduced in zopolrestat treated diabetic and non-diabetic hearts (P = 0.005). Zopolrestat increased the activity of Na-,K(+)-ATPase in diabetic hearts under baseline conditions (11.70 +/- 0.95 versus 7.28 +/- 0.98 mumol/h/mg protein, P = 0.005) as well as during ischemia and reperfusion. Similar changes in Na+,K(+)-ATPase activity were also observed in non-diabetic hearts. CONCLUSIONS: The data provide additional support to the protective effects of zopolrestat and suggest that a possible mechanism of action may be associated with the attenuation of the rise in [Na]i and [Ca]i during ischemia and reperfusion.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Metals/metabolism , Myocardial Ischemia/drug therapy , Myocardium/metabolism , Phthalazines/therapeutic use , Thiazoles/therapeutic use , Analysis of Variance , Animals , Benzothiazoles , Calcium/analysis , Calcium/metabolism , Diabetes Mellitus, Type 2/metabolism , Enzyme Inhibitors/metabolism , Intracellular Fluid/chemistry , Intracellular Fluid/metabolism , Magnetic Resonance Spectroscopy , Metals/analysis , Myocardial Ischemia/metabolism , Myocardial Reperfusion , Perfusion , Rats , Rats, Inbred BB , Sodium/analysis , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Time Factors
8.
Diabetes ; 46(2): 292-300, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9000707

ABSTRACT

Diabetes increases the incidence of cardiovascular disease as well as the complications of myocardial infarction. Studies using animal models of diabetes have demonstrated that the metabolic alterations occurring at the myocyte level may contribute to the severity of ischemic injury in diabetic hearts. Of the several mechanisms being investigated to understand the pathogenesis of diabetic complications, the increased metabolism of glucose via the polyol pathway has received considerable attention. Deviant metabolic regulation due to increased flux through aldose reductase in diabetic hearts may influence the ability of the myocardium to withstand ischemia insult. To determine if aldose reductase inhibition improves tolerance to ischemia, hearts from acute type I diabetic and nondiabetic control rats were isolated and retrograde perfused. Each group was exposed to 1 micromol/l zopolrestat, a specific inhibitor of aldose reductase, for 10 min, followed by 20 min of global ischemia and 60 min of reperfusion in the absence of zopolrestat. Zopolrestat reduced sorbitol levels before ischemia in diabetic hearts. The cytosolic redox state (NADH/NAD+), as measured by lactate-to-pyruvate ratios, was significantly lowered under baseline, ischemic, and reperfusion conditions in diabetic hearts perfused with zopolrestat. In these diabetic hearts, ATP was significantly higher in zopolrestat hearts during ischemia, as were phosphocreatine and left ventricular-developed pressure on reperfusion. Zopolrestat provided similar metabolic and functional benefits in nondiabetic hearts. Creatine kinase release was reduced by approximately 50% in both nondiabetic and diabetic hearts treated with zopolrestat. These data indicate that inhibition of aldose reductase activity preserves high-energy phosphates, maintains a lower cytosolic NADH/NAD+ ratio, and markedly protects both diabetic and nondiabetic hearts during ischemia and reperfusion.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Diabetes Mellitus, Type 1/physiopathology , Myocardial Ischemia/physiopathology , Phthalazines/pharmacology , Reperfusion Injury/prevention & control , Thiazoles/pharmacology , Adenosine Triphosphate/metabolism , Animals , Benzothiazoles , Blood Glucose/metabolism , Energy Metabolism , Enzyme Inhibitors/pharmacology , Fructose/metabolism , Hydrogen-Ion Concentration , Lactates/metabolism , Magnetic Resonance Spectroscopy , Myocardium/metabolism , NAD/metabolism , Pyruvates/metabolism , Rats , Rats, Inbred BB , Reperfusion Injury/physiopathology , Sorbitol/metabolism
9.
Diabetes ; 43(9): 1122-9, 1994 Sep.
Article in English | MEDLINE | ID: mdl-8070612

ABSTRACT

Similar vascular pathological conditions are observed in diabetic animals and those with diet-induced hypergalactosemia. Both diabetes and hypergalactosemia are believed to cause vascular dysfunction via a common biochemical mechanism. In this study, we have found that both diabetes and hypergalactosemia in the short term (2-4 months) can increase total diacylglycerol (DAG) levels by 52 +/- 9 and 74 +/- 13% in the retina and aorta, respectively, of diabetic dogs, and by 94 +/- 9 and 78 +/- 11% in the retina and aorta, respectively, in dogs with hypergalactosemia as compared with normal control animals (P < 0.01). The elevation of DAG levels was maintained for 5 years in the aortas of diabetic and hypergalactosemic dogs. To characterize the mechanism of the DAG increases, we have determined that total DAG levels were significantly increased in cultured macro- and microvascular cells exposed to elevated glucose (22 mM) and galactose (16.5 mM) levels. These increased levels were not prevented by sorbinil, an aldose reductase inhibitor. One of the sources of the increased DAG levels was probably derived from de novo synthesis from both hexoses as determined by radiolabeling studies. Intracellularly, the DAG elevation activated protein kinase C (PKC) activity with increases of 58 +/- 12% (P < 0.05) and 66 +/- 8% (P < 0.01) in the membrane fraction of cultured aortic smooth muscle cells exposed to elevated glucose and galactose levels, respectively. These findings have clearly demonstrated a possible common biochemical mechanism by which hyperglycemia and hypergalactosemia can chronically activate the DAG-PKC pathway in the vasculature and could be a possible explanation for the development of diabetic vascular complications.


Subject(s)
Aorta/metabolism , Diabetes Mellitus, Experimental/metabolism , Diglycerides/metabolism , Endothelium, Vascular/metabolism , Galactosemias/metabolism , Muscle, Smooth, Vascular/metabolism , Protein Kinase C/metabolism , Retina/metabolism , Animals , Aorta/drug effects , Arachidonic Acid/metabolism , Cell Membrane/enzymology , Cells, Cultured , Cytosol/enzymology , Dogs , Endothelium, Vascular/drug effects , Female , Galactose/metabolism , Galactose/pharmacology , Glucose/metabolism , Glucose/pharmacology , Kinetics , Male , Muscle, Smooth, Vascular/drug effects , Palmitic Acid , Palmitic Acids/metabolism , Phosphatidylcholines/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Reference Values , Retina/drug effects , Tritium
10.
Metabolism ; 43(4): 492-500, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8159110

ABSTRACT

These experiments were undertaken to assess the effects of combined treatment with insulin (designed to partially restore metabolic control) and sorbinil (an aldose reductase inhibitor [ARI]) versus the effects of sorbinil alone or of two insulin regimens providing different degrees of glycemic control on diabetes-induced metabolic derangements and vascular function. Streptozocin-diabetic rats were divided into the following five groups: (1) untreated (D); (2) treated with approximately 1 U NPH insulin/100 g body weight/d administered in one subcutaneous (SC) injection (DI-1); (3) treated with the same total daily dose of insulin administered in two SC injections (DI-2); (4) treated with approximately 0.2 mmol sorbinil in the diet/kg body weight/d (DS); and (5) treated with once-daily insulin plus sorbinil (DSI-1). Two groups of nondiabetic rats, untreated (C) and sorbinil-treated (CS), served as controls. Metabolic parameters were unaffected by sorbinil treatment in controls and diabetics, whereas insulin administration in the diabetics virtually normalized body growth, food consumption, urine volume, and plasma glucose levels, and markedly decreased hemoglobin A1 (HbA1) levels. Two daily injections were more effective than one in improving metabolic control as measured by HbA1 levels. Regional vascular 131I-albumin permeation was increased about twofold to threefold by diabetes in ocular tissues, sciatic nerve, aorta, diaphragm, and new granulation tissue; it was decreased (but not normalized) by insulin treatment in accordance with improved metabolic control, and was completely normalized by sorbinil. 131I-albumin kidney clearance, as well as urinary albumin and IgG excretion, were markedly increased in diabetic rats and were significantly decreased but not completely normalized by sorbinil and by twice-daily insulin treatment.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Angiopathies/drug therapy , Imidazoles/therapeutic use , Imidazolidines , Insulin/therapeutic use , Aldehyde Reductase/antagonists & inhibitors , Animals , Body Weight , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetic Angiopathies/physiopathology , Diabetic Angiopathies/prevention & control , Drug Therapy, Combination , Kidney/physiopathology , Male , Organ Size , Rats , Rats, Sprague-Dawley , Serum Albumin, Bovine/metabolism
11.
Am J Physiol ; 265(4 Pt 2): H1189-94, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8238405

ABSTRACT

This study addresses the question of whether a decrease in basal Na+ pump [Na(+)-K(+)-adenosinetriphosphatase (ATPase)] activity occurs in the carotid artery of an alloxan-diabetic rabbit and, if so, whether it is associated with altered 86Rb+ uptake and contractile response to ouabain and K(+)-free solution. Ouabain-sensitive 86Rb+ uptake, an index of Na+ pump activity, was diminished approximately 50% in carotid arteries from diabetic rabbits. Concurrent with this, contractions induced by incubating the carotid arteries in a K(+)-free solution (in the absence of phentolamine) were significantly larger in the diabetic group. Readdition of K+ (1 mM) to arteries contracted with the K(+)-free solution caused relaxations that were slower to occur and of lesser magnitude in diabetic than in control rabbits. In contrast to the contractions caused by the K(+)-free medium, contractions caused by incubation with ouabain (1 mM) in the presence of phentolamine were significantly smaller in the diabetic group. Treatment of diabetic rabbits with an aldose reductase inhibitor, zopolrestat, at both high and low doses restored the alterations in vascular reactivity toward normal. The results indicate that the Na+ pump activity is diminished in the carotid artery of diabetic rabbit, and this is associated with abnormal vascular responsiveness and increased polyol pathway flux.


Subject(s)
Carotid Arteries/metabolism , Diabetes Mellitus, Experimental/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Aldehyde Reductase/antagonists & inhibitors , Animals , Benzothiazoles , In Vitro Techniques , Male , Ouabain/pharmacology , Phthalazines/pharmacology , Potassium/pharmacology , Rabbits , Rubidium/pharmacokinetics , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Thiazoles/pharmacology , Vasoconstriction/drug effects , Vasodilation/drug effects
13.
Am J Obstet Gynecol ; 167(6): 1837-43, 1992 Dec.
Article in English | MEDLINE | ID: mdl-1471707

ABSTRACT

OBJECTIVE: Our objective was to determine whether impaired ovarian function induced by short-term creation of a galactosemic state in the rat might be prevented by the coadministration of an aldose reductase inhibitor. STUDY DESIGN: Prepubertal Sprague-Dawley rats were fed four different diets including (1) control, (2) 40% galactose, (3) 40% galactose and an aldose reductase inhibitor, and (4) an aldose reductase inhibitor with the control diet. Percentage germinal vesicle breakdown, postovulatory oocyte quantities, hormonal parameters, ovarian histologic evaluation, and ovarian galactitol concentrations were determined. RESULTS: The galactose-fed animals (group 2) had decreased germinal vesicle breakdown (47%) versus control (69%, p < 0.05). Galactose-exposed animals had significantly decreased quantities of postovulatory eggs (6.4 per animal) after menotropin ovarian stimulation in comparison with controls (14.1, p < 0.01). In rats exposed to high dietary levels of galactose (group 2) ovarian galactitol concentrations were significantly higher (protein 42.12 mumol/gm versus 0.0 for controls, p < 0.005). When galactose-fed animals received the aldose reductase inhibitor, ovarian accumulation of galactitol was significantly reduced and the observed detrimental effects on the oocyte were prevented. CONCLUSION: Galactitol accumulation or metabolic flux through aldose reductase in galactosemic rodents may be involved in the demonstrated ovarian dysfunction.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Galactose , Ovarian Diseases/prevention & control , Animals , Female , Galactitol/metabolism , Galactose/pharmacology , Granulosa Cells/pathology , Ovarian Diseases/chemically induced , Ovarian Diseases/pathology , Ovary/metabolism , Ovary/pathology , Rats , Rats, Sprague-Dawley
14.
Dig Dis Sci ; 36(12): 1721-8, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1748041

ABSTRACT

CP-66,948 is a histamine H2-receptor antagonist with gastric antisecretory activity and mucosal protective properties. The affinity of CP-66,948 for the guinea pig atria histamine H2-receptor is 15 times greater than that of cimetidine and seven times greater than that of ranitidine. In vivo, the ED50 value for inhibition of gastric acid secretion in pylorus-ligated rats is 2 mg/kg intraduodenally, and in histamine or pentagastrin-stimulated Heidenhain pouch dogs the antisecretory ED50 values are 0.3 mg/kg per os and 1.0 mg/kg per os, respectively. CP-66,948 also inhibits ethanol-induced gastric hemorrhagic lesions in rats following either oral or systemic administration (ED50 values of 12 mg/kg per os and 6 mg/kg subcutaneously). In addition, the mucosal protective activity is independent of prostaglandin synthesis. CP-66,948 inhibits gastric acid secretion in man, and its mucosal protective activity may provide additional benefits in peptic ulcer therapy.


Subject(s)
Gastric Mucosa/drug effects , Imidazoles/pharmacology , Thiazoles/pharmacology , Animals , Cimetidine/pharmacology , Dogs , Dose-Response Relationship, Drug , Ethanol/toxicity , Gastric Acid/metabolism , Gastric Mucosa/pathology , Gastrointestinal Hemorrhage/chemically induced , Gastrointestinal Hemorrhage/pathology , Guinea Pigs , Heart Rate/drug effects , Histamine/pharmacology , In Vitro Techniques , Indomethacin/pharmacology , Male , Pentagastrin/pharmacology , Ranitidine/pharmacology , Rats , Rats, Inbred Strains , Time Factors
15.
Gastroenterology ; 94(1): 10-21, 1988 Jan.
Article in English | MEDLINE | ID: mdl-3335281

ABSTRACT

Concentrated ethanol causes gastric lesions by a mechanism that is poorly understood. We have investigated this mechanism in the rat stomach via gross morphologic, videomicroscopic, histochemical, and pharmacologic approaches. Within 1 min of contact, ethanol caused diffuse mucosal hyperemia. By 5 min, hyperemia greatly intensified at some mucosal sites. Beneath sites where mucosal hyperemia developed, intramural venules strongly constricted at 3-13 s postethanol, whereas submucosal arterioles dilated more than two times in diameter by 25 s. Submucosal venular constriction began sooner than arteriolar dilation (9 vs. 16 s, p less than 0.05). One-third of the gastric mucosal mast cells degranulated by 15 s postethanol; 50% discharged by 30 s. Ethanol-induced hyperemia was markedly reduced by lipoxygenase-selective inhibitors BW755C or nordihydroguaiaretic acid, or by the H1-antihistamine pyrilamine, but not by indomethacin, cimetidine, phentolamine, or methysergide. Based on these results, a model for the pathogenesis of ethanol-induced gastric lesions is proposed.


Subject(s)
Ethanol/toxicity , Gastric Mucosa/drug effects , Hyperemia/chemically induced , Animals , Gastric Mucosa/blood supply , Male , Mast Cells/pathology , Rats , Vasoconstriction/drug effects , Vasodilation/drug effects , Videotape Recording
16.
J Med Chem ; 29(11): 2154-63, 1986 Nov.
Article in English | MEDLINE | ID: mdl-2878075

ABSTRACT

The structural relationship of the competitive histamine H2-receptor antagonist 3-amino-5-(2-amino-4-pyridyl)-1,2,4-triazole (1) to the agonist histamine and to antagonists of the cimetidine type was explored by the design and synthesis of four series of bioisosterically designed prototypes. Biological data from these series was best interpreted as indicating a similarity between the imidazole moiety of histamine and cimetidine and the 2-amino-4-pyridyl moiety of 1. On the basis of this data, sequential replacement of 2-amino-4-pyridyl by 2-[(dimethylamino)methyl]-5-furyl and 2-guanidino-4-triazolyl moieties led to a structurally more potent series of biaryl histamine H2-receptor antagonists. The best of these, 2-methyl-4-(2-guanidino-4-thiazolyl)imidazole (29, CP-57,361-1) was 120 times more potent as a histamine H2-receptor antagonist than 1.


Subject(s)
Histamine H2 Antagonists/pharmacology , Imidazoles/pharmacology , Triazoles/pharmacology , Animals , Binding, Competitive , Cimetidine/pharmacology , Dogs , Guinea Pigs , Histamine/pharmacology , Histamine H2 Antagonists/chemical synthesis , In Vitro Techniques , Pyridines/pharmacology , Structure-Activity Relationship
17.
J Cell Biol ; 85(3): 804-10, 1980 Jun.
Article in English | MEDLINE | ID: mdl-6248567

ABSTRACT

Fusion of phagolysosomes has been previously demonstrated to occur during the incubation of phagolysosome-containing homogenates of Acanthamoeba (Oates and Touster, 1978, J. Cell Biol. 79:217-234). Further studies on this system have shown that methylxanthines (0.2 mM) and/or cAMP (0.5-1 mM) markedly accelerate the average rate, but not the extent, of the in vitro phagolysosome fusion process. Adenosine, 5'-AMP, and ADP (0.5-1 mM) were without effect. ATP (0.5-1 mM) caused variable stimulation, whereas beta, gamma-methylene-ATP (1 mM) caused pronounced inhibition, as did GTP (1 mM) and cGMP (1 mM). Stimulation by 3-isobutyl-1-methylxanthine was blocked by GTP, but not by ATP or cAMP. These results indicate that the rate of phagolysosome fusion in Acanthamoeba homogenates may be regulated by cyclic nucleotides, with enhancement of the fusion rate by cAMP and inhibition of the rate by cGMP. The extent of the reaction increased spontaneously and markedly during the first few hours after preparation of the homogenates. This activation appears to be because of a slow conversion of a significant fraction of the vacuole population from a fusion-incompetent to a fusion-competent, cyclic nucleotide-sensitive state.


Subject(s)
Amoeba/ultrastructure , Intracellular Membranes/ultrastructure , Lysosomes/ultrastructure , Nucleotides, Cyclic/pharmacology , Organoids/ultrastructure , Vacuoles/ultrastructure , Adenosine Triphosphate/pharmacology , Animals , Cell-Free System , Cyclic AMP/pharmacology , Guanosine Triphosphate/pharmacology , Intracellular Membranes/drug effects , Phosphodiesterase Inhibitors/pharmacology , Time Factors
18.
J Cell Biol ; 79(1): 217-34, 1978 Oct.
Article in English | MEDLINE | ID: mdl-701373

ABSTRACT

To investigate the properties of phagolysosome (PL) fusion in Acanthamoeba homogenates, it was necessary to develop reliable methods for measuring in vitro PL fusion. The need to distinguish PL fusion from PL adhesion was met by the development of a quantitative electron microscope assay. Initial characterization of the fusion reaction by this method was followed by the development of a more rapid light microscope assay. Results obtained by the two methods were found to be in close agreement. By use of these new techniques, the in vitro PL fusion reaction was demonstrated to occur in a quantitatively reproducible manner. Under the present conditions employed, PL breakdown was not detected at any time during the in vitro incubation, while PL fusion was observed to proceed linearly for approximately 10 min, at which time the reaction ceased. Incubation of mixtures of two distinct PL types resulted in increases in hybrid PL types that were paralleled by decreases in nonhybrid PL types. The relative changes in PL concentrations observed were quantitatively consistent with PL fusion occurring randomly with respect to PL type. PL fusion was strongly inhibited by low concentrations of KF (50% inhibition at 2.7 mM), and by approximately tenfold higher concentrations of KCl, while KCN and 2,4-dinitrophenol (2,4-DNP) had little effect. In addition to further defining the nature of the PL fusion reaction in this system, these results demonstrate that, by use of the techniques described, quantitative study of the biochemical properties of this reaction is now possible.


Subject(s)
Amoeba/ultrastructure , Lysosomes/physiology , Animals , Dinitrophenols/pharmacology , Fluorides/pharmacology , Lysosomes/drug effects , Lysosomes/ultrastructure , Mathematics , Microscopy, Electron , Potassium Chloride/pharmacology
19.
J Cell Biol ; 68(2): 319-38, 1976 Feb.
Article in English | MEDLINE | ID: mdl-1245550

ABSTRACT

Fusion of phagolysosomes (PLs) has been demonstrated to occur in vitro. Two separate cell homogenates of the ameba Acanthamoeba sp. (Neff) were prepared, each rich in PLs labeled with distinctive particulate markers. Portions of each were incubated together in vitro and fusion occurred as evidenced by the appearance of PLs containing both types of markers. Fusion was confirmed by electron microscopy, including serial sectioning. The membranes of fused vacuoles excluded the dye eosin Y. Surviving cells in the homogenates were not responsible for the observed fusion. Fusion was obtained using either synthetic markers (polystyrene and polyvinyltoluene latex) or biological markers (autoclaved yeast cells and glutaraldehyde-fixed goat red blood cells), or a combination of both. The specificity of PL fusion in vivo appeared to be maintained in vitro. As determined by light and electron microscopy, the fusion reaction was dependent on time and temperature, and on the initial presence of membrane around both marker particles. A minimum of 10% of the vacuoles fused by 10 min of incubation at 30 degrees C, and no rupture of the vacuoles was detected during this time. After 10 min of incubation, vacuole rupture began and fusion ceased. At a constant initial vacuole concentration, the extent of PL fusion in vitro was quantitatively reproducible. This appears to be a promising system for further investigation of membrane fusion in the lysosomal system.


Subject(s)
Amoeba/physiology , Lysosomes/physiology , Phagocytosis , Amoeba/ultrastructure , Animals , Dinitrophenols/pharmacology , Erythrocytes , Kinetics , Latex , Membranes/physiology , Membranes/ultrastructure , Microspheres , Phagocytosis/drug effects , Temperature , Yeasts
SELECTION OF CITATIONS
SEARCH DETAIL
...