Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Curr Oncol ; 18(1): 11-2, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21331275
3.
Am Heart J ; 135(5 Pt 1): 914-23, 1998 May.
Article in English | MEDLINE | ID: mdl-9588425

ABSTRACT

BACKGROUND: The natriuretic peptide family consists of four molecules that share significant amino acid sequence homologic characteristics and a looped motif. Atrial natriuretic peptide and brain natriuretic peptide are similar in their ability to promote natriuresis and diuresis, inhibit the renin-angiotensin-aldosterone axis, and act as vasodilators. Understanding of the actions of C-type natriuretic peptide and dendroaspis natriuretic peptide is incomplete, but these two new family members also act as vasodilators. Because of the rapid evolution of information about this peptide family, we reviewed the state of the art with respect to risk stratification and therapeutic ability. METHODS: English-language papers were identified by a MEDLINE database search covering 1966 through 1997 and supplemented with bibliographic references and texts. CONCLUSIONS: The natriuretic peptides are counterregulatory hormones with prognostically important levels. They are similarly upregulated in heart failure and counteract neurohormones that induce vasoconstriction and fluid retention. BNP may be the superior prognosticator for risk stratification after myocardial infarction and is independent of left ventricular ejection fraction. Lastly, experimental trials suggest that administration of exogenous natriuretic peptides or inhibitors of their catabolism to patients with ischemic heart disease may be clinically beneficial.


Subject(s)
Atrial Natriuretic Factor/physiology , Myocardial Ischemia/physiopathology , Animals , Atrial Natriuretic Factor/therapeutic use , Humans , Myocardial Infarction/diagnosis , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Myocardial Ischemia/diagnosis , Myocardial Ischemia/therapy , Vasodilation/drug effects , Vasodilation/physiology
4.
J Exp Med ; 186(9): 1615-20, 1997 Nov 03.
Article in English | MEDLINE | ID: mdl-9348321

ABSTRACT

The complete healing of wounds is the final step in a highly regulated response to injury. Although many of the molecular mediators and cellular events of healing are known, their manipulation for the enhancement and acceleration of wound closure has not proven practical as yet. We and others have established that adenosine is a potent regulator of the inflammatory response, which is a component of wound healing. We now report that ligation of the G alpha s-linked adenosine receptors on the cells of an artificial wound dramatically alters the kinetics of wound closure. Excisional wound closure in normal, healthy mice was significantly accelerated by topical application of the specific A2A receptor agonist CGS-21680 (50% closure by day 2 in A2 receptor antagonists. In rats rendered diabetic (streptozotocin-induced diabetes mellitus) wound healing was impaired as compared to nondiabetic rats; CGS-21680 significantly increased the rate of wound healing in both nondiabetic and diabetic rats. Indeed, the rate of wound healing in the CGS-21680-treated diabetic rats was greater than or equal to that observed in untreated normal rats. These results appear to constitute the first evidence that a small molecule, such as an adenosine receptor agonist, accelerates wound healing in both normal animals and in animals with impaired wound healing.


Subject(s)
Adenosine/analogs & derivatives , Phenethylamines/administration & dosage , Purinergic P1 Receptor Agonists , Wound Healing/drug effects , Adenosine/administration & dosage , Administration, Topical , Animals , Cell Line , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Endothelium, Vascular/metabolism , Female , Fibroblasts/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A2A , Receptors, Purinergic P1/biosynthesis , Receptors, Purinergic P1/genetics , Skin , Umbilical Veins
5.
Am J Med Qual ; 12(3): 169-74, 1997.
Article in English | MEDLINE | ID: mdl-9287457

ABSTRACT

We report on the introduction of a new technology and a new method for the management of chronic coronary artery disease into a managed care environment. The introduction incurred substantial resistance from subspecialty consultants, primary care physicians, and top management. Strategies were developed to overcome these resistances. Modification of the program as well as the development of incentives occurred. These measures continue to evolve. The program, to date, has achieved approximately 50% penetration. The demonstration of better health outcomes and financial savings will almost certainly temper the resistance encountered from all three groups identified.


Subject(s)
Disease Management , Electrocardiography, Ambulatory/statistics & numerical data , Managed Care Programs/organization & administration , Myocardial Ischemia/diagnosis , Myocardial Ischemia/therapy , Technology Transfer , Attitude of Health Personnel , California , Cost Savings , Diffusion of Innovation , Humans , Models, Organizational , Outcome Assessment, Health Care
6.
J Immunol ; 156(5): 1937-41, 1996 Mar 01.
Article in English | MEDLINE | ID: mdl-8596047

ABSTRACT

The anti-inflammatory mechanism of sulfasalazine is not well understood. It has recently been shown that sulfasalazine inhibits 5-aminoimidazole-4-carboxamidoribonucleotide (AICAR) transformylase, an enzyme involved in de novo purine biosynthesis. We recently demonstrated that methotrexate promotes intracellular AICAR accumulation, thereby increasing adenosine release and diminishing inflammation, so we tested the hypothesis that sulfasalazine similarly promotes intracellular AICAR accumulation. We studied adenosine release and the state of inflammation in in vitro and in vivo models of the inflammatory process. The adhesion of stimulated neutrophils (FMLP) to endothelial cells preincubated with sulfasalazine was inhibited in a dose-dependent manner. Elimination of extracellular adenosine by addition of adenosine deaminase or inhibition of adenosine by the adenosine A2 receptor antagonist 3,7-dimethyl-1-propargylxanthine (DMPX) completely reversed the anti-inflammatory effect of sulfasalazine (at concentrations <1 microM in this in vitro model. To determine whether this phenomenon was relevant to inhibition of inflammation in vivo, we studied the effect of sulfasalazine (100 mg/kg/day by gastric gavage for 3 days) on leukocyte accumulation in the murine air pouch model of inflammation. Treatment with sulfasalazine markedly decreased the number of leukocytes that accumulated in the inflamed (carrageenan, 2 mg/ml) air pouch. Injection of either adenosine deaminase or DMPX, but not the A1 receptor antagonist 8-cyclopentyl-dipropylxanthine, significantly reversed the anti-inflammatory effects of sulfasalazine treatment. Sulfasalazine increased the exudate adenosine concentration from 127 +/- 64 nM to 869 +/- 47 nM. Moreover, sulfasalazine treatment promoted a marked increase in splenocyte AICAR concentration from 35 +/- 6 to 96 +/- 3 pmols/10(6) splenocytes, which is consistent with the in vitro observation that sulfasalazine inhibits AICAR transformylase. These results indicate that sulfasalazine, like methotrexate, enhances adenosine release at an inflamed site and that adenosine diminishes inflammation via occupancy of A2 receptors on inflammatory cells. Our studies provide evidence that sulfasalazine and methotrexate may be described as a newly recognized family of anti-inflammatory agents that share the property of using adenosine as an antagonist of inflammation.


Subject(s)
Adenosine/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Endothelium, Vascular/pathology , Hydroxymethyl and Formyl Transferases , Sulfasalazine/pharmacology , Acyltransferases/antagonists & inhibitors , Adenosine/physiology , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/metabolism , Animals , Carrageenan/toxicity , Disease Models, Animal , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Neutrophil Activation/drug effects , Phosphoribosylaminoimidazolecarboxamide Formyltransferase , Ribonucleotides/metabolism , Sulfasalazine/administration & dosage
7.
Hypertension ; 27(2): 245-50, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8567047

ABSTRACT

The objective of this study was to determine whether the calcium antagonist amlodipine could slow the progression of chronic renal disease. We examined the effects of amlodipine on kidney structure and function in two experimental models of hypertension. In the first study, adult, male Munich Wistar rats underwent uninephrectomy and were given weekly injections of desoxycorticosterone and 1% saline for drinking. Rats ingested normal chow or chow containing amlodipine for 8 weeks. The drug reduced systemic blood pressure, but glomerular filtration rate, kidney weight, proteinuria, and morphological evidence of glomerular injury were not affected. In the second study, male spontaneously hypertensive rats underwent uninephrectomy at 5 weeks of age and were followed for 6 months, during which they received no therapy or amlodipine. The drug dose was determined in preliminary studies to be the highest dose not associated with marked growth retardation. Again, although systemic blood pressure was significantly reduced by amlodipine, proteinuria and the prevalence of glomerulosclerosis were similar in amlodipine-treated and control spontaneously hypertensive rats. Micropuncture studies revealed that glomerular pressure remained elevated in amlodipine-treated spontaneously hypertensive rats. Kidney weight and glomerular volume were also similar in amlodipine-treated and control rats. Amlodipine also failed to inhibit platelet aggregation. Therefore, antihypertensive therapy with amlodipine fails to reduce glomerular pressure in spontaneously hypertensive rats as well as glomerular size and injury in spontaneously hypertension rats and desoxycorticosterone-salt hypertension. Although other dihydropyridine calcium antagonists have been found to reduce experimental glomerular injury, these data suggest that amlodipine may not prevent hypertensive nephrosclerosis.


Subject(s)
Amlodipine/pharmacology , Antihypertensive Agents/pharmacology , Glomerular Filtration Rate/drug effects , Hypertension, Renal/physiopathology , Kidney Glomerulus/drug effects , Animals , Blood Pressure/drug effects , Body Weight/drug effects , Desoxycorticosterone , Kidney/drug effects , Kidney Glomerulus/pathology , Kidney Glomerulus/physiopathology , Male , Nephrectomy , Organ Size/drug effects , Platelet Aggregation/drug effects , Proteinuria , Rats , Rats, Wistar
8.
J Clin Invest ; 96(2): 994-1002, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7543498

ABSTRACT

Since colchicine-sensitive microtubules regulate the expression and topography of surface glycoproteins on a variety of cells, we sought evidence that colchicine interferes with neutrophil-endothelial interactions by altering the number and/or distribution of selectins on endothelial cells and neutrophils. Extremely low, prophylactic, concentrations of colchicine (IC50 = 3 nM) eliminated the E-selectin-mediated increment in endothelial adhesiveness for neutrophils in response to IL-1 (P < 0.001) or TNF alpha (P < 0.001) by changing the distribution, but not the number, of E-selectin molecules on the surface of the endothelial cells. Colchicine inhibited stimulated endothelial adhesiveness via its effects on microtubules since vinblastine, an agent which perturbs microtubule function by other mechanisms, diminished adhesiveness whereas the photoinactivated colchicine derivative gamma-lumicolchicine was inactive. Colchicine had no effect on cell viability. At higher, therapeutic, concentrations colchicine (IC50 = 300 nM, P < 0.001) also diminished the expression of L-selectin on the surface of neutrophils (but not lymphocytes) without affecting expression of the beta 2-integrin CD11b/CD18. In confirmation, L-selectin expression was strikingly reduced (relative to CD11b/CD18 expression) on neutrophils from two individuals who had ingested therapeutic doses of colchicine. These results suggest that colchicine may exert its prophylactic effects on cytokine-provoked inflammation by diminishing the qualitative expression of E-selectin on endothelium, and its therapeutic effects by diminishing the quantitative expression of L-selectin on neutrophils.


Subject(s)
Cell Adhesion Molecules/biosynthesis , Colchicine/pharmacology , Endothelium, Vascular/drug effects , Neutrophils/drug effects , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , CD18 Antigens/biosynthesis , Cell Adhesion/drug effects , Cells, Cultured , E-Selectin , Endothelium, Vascular/metabolism , Endothelium, Vascular/ultrastructure , Humans , Interleukin-1/pharmacology , L-Selectin , Macrophage-1 Antigen/biosynthesis , Mice , Microtubules/drug effects , Neutrophils/metabolism , Neutrophils/ultrastructure , Tumor Necrosis Factor-alpha/pharmacology , Umbilical Veins , Vinblastine/pharmacology
9.
Proc Natl Acad Sci U S A ; 92(18): 8378-82, 1995 Aug 29.
Article in English | MEDLINE | ID: mdl-7545301

ABSTRACT

To examine the role of complement components as regulators of the expression of endothelial adhesive molecules in response to immune complexes (ICs), we determined whether ICs stimulate both endothelial adhesiveness for leukocytes and expression of E-selectin and intercellular and vascular cell adhesion molecules 1 (ICAM-1 and VCAM-1). We found that ICs [bovine serum albumin (BSA)-anti-BSA] stimulated endothelial cell adhesiveness for added leukocytes in the presence of complement-sufficient normal human serum (NHS) but not in the presence of heat-inactivated serum (HIS) or in tissue culture medium alone. Depletion of complement component C3 or C8 from serum did not prevent enhanced endothelial adhesiveness stimulated by ICs. In contrast, depletion of complement component C1q markedly inhibited IC-stimulated endothelial adhesiveness for leukocytes. When the heat-labile complement component C1q was added to HIS, the capacity of ICs to stimulate endothelial adhesiveness for leukocytes was completely restored. Further evidence for the possible role of C1q in mediating the effect of ICs on endothelial cells was the discovery of the presence of the 100- to 126-kDa C1q-binding protein on the surface of endothelial cells (by cytofluorography) and of message for the 33-kDa C1q receptor in resting endothelial cells (by reverse transcription-PCR). Inhibition of protein synthesis by cycloheximide blocked endothelial adhesiveness for leukocytes stimulated by either interleukin 1 or ICs in the presence of NHS. After stimulation with ICs in the presence of NHS, endothelial cells expressed increased numbers of adhesion molecules (E-selectin, ICAM-1, and VCAM-1). Endothelial expression of adhesion molecules mediated, at least in part, endothelial adhesiveness for leukocytes, since leukocyte adhesion was blocked by monoclonal antibodies directed against E-selectin. These studies show that ICs stimulate endothelial cells to express adhesive proteins for leukocytes in the presence of a heat-labile serum factor. That factor appears to be C1q.


Subject(s)
Antigen-Antibody Complex/metabolism , Cell Adhesion Molecules/biosynthesis , Complement C1q/metabolism , Endothelium, Vascular/metabolism , Base Sequence , Cells, Cultured , DNA Primers , E-Selectin , Endothelium, Vascular/cytology , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Leukocytes/cytology , Molecular Sequence Data , Neutrophils/cytology , Polymerase Chain Reaction , Vascular Cell Adhesion Molecule-1
10.
Inflammation ; 18(3): 323-35, 1994 Jun.
Article in English | MEDLINE | ID: mdl-8088928

ABSTRACT

All nonsteroidal antiinflammatory drugs (NSAIDs) inhibit neutrophil aggregation (homotypic cell-cell adhesion) and do so without affecting expression of CD11b/CD18. Since the first step in acute inflammation is a critical interaction between neutrophils and the vascular endothelium (heterotypic cell-cell adhesion), we determined whether NSAIDs diminish the adherence of neutrophils to the endothelium. At antiinflammatory concentrations (0.5-5 mM) sodium salicylate, an NSAID that does not inhibit prostaglandin synthesis, inhibited stimulated but not unstimulated neutrophil adherence to endothelial cells (IC50 < 1 mM, P < 0.00001). Salicylates have previously been shown to inhibit oxidative phosphorylation and, predictably, sodium salicylate inhibited oxidative phosphorylation, as evidenced by depletion of ATP stores (875 +/- 75 pmol/10(6) PMN, [2.92 +/- 0.25 mM]) in stimulated (FMLP, 0.1 microM) but not resting neutrophils treated with antiinflammatory doses of sodium salicylate (EC50 = 1 mM, P < 0.00001). Indomethacin and piroxicam (10 and 30 microM) only minimally decreased ATP concentrations in stimulated and resting neutrophils. ATP is metabolized to adenosine, and we have previously demonstrated that both endogenously released (180-200 nM) and exogenous adenosine (IC50 = 250 nM) inhibit stimulated neutrophil adherence to endothelial cells. To determine whether the increased metabolism of ATP and the resultant increase in adenosine release were responsible for inhibition of neutrophil adhesion to endothelium, we determined whether addition of adenosine deaminase (ADA, 0.125 IU/ml), an enzyme that converts extracellular adenosine to its inactive metabolite, inosine, affected inhibition of neutrophil adhesion to endothelium by stimulated neutrophils. ADA significantly reversed inhibition of neutrophil adherence to endothelium by sodium salicylate (0.5-5 mM, P < 0.00001). This suggests that sodium salicylate inhibits neutrophil adherence by increasing adenosine release. Whereas indomethacin and piroxicam (10-50 microM) also inhibited stimulated neutrophil adherence to endothelial cells, ADA did not affect their inhibition of adherence. These studies demonstrate a heretofore unexpected antiinflammatory mechanism for salicylates: salicylates increase ATP hydrolysis and thereby enhance release of adenosine. Moreover, these data are consistent with the hypothesis that NSAIDs differ from one another with respect to their mechanisms of action.


Subject(s)
Adenosine/physiology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Endothelium, Vascular/physiology , Neutrophils/drug effects , Adenosine/metabolism , Adenosine Triphosphate/metabolism , Cell Adhesion/drug effects , Humans , Indomethacin/pharmacology , Neutrophils/metabolism , Neutrophils/physiology , Osmolar Concentration , Piroxicam/pharmacology , Sodium Salicylate/pharmacology
11.
Med Hypotheses ; 40(3): 169-73, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8502196

ABSTRACT

Duchenne's muscular dystrophy (DMD) is a common X-linked neuromuscular disease which predominantly affects skeletal and cardiac muscle. The absence of dystrophin, the metabolic defect that causes DMD, leads to a peculiar cardiomyopathy which initially affects the posterior wall of the left ventricle. We review evidence that dystrophin deficient myocytes become dystrophic in order of increasing axial stress upon the myocyte. Thus, dystrophin's function may be that of physically reinforcing the sarcolemma against the axial forces exerted upon the myocyte.


Subject(s)
Cardiomyopathies/etiology , Dystrophin/physiology , Heart/anatomy & histology , Muscular Dystrophies/physiopathology , Animals , Cardiomyopathies/physiopathology , Dogs , Dystrophin/genetics , Heart/physiology , Heart/physiopathology , Humans , Muscular Dystrophies/genetics , Muscular Dystrophy, Animal/physiopathology , X Chromosome
12.
Proc Natl Acad Sci U S A ; 89(21): 9991-5, 1992 Nov 01.
Article in English | MEDLINE | ID: mdl-1279685

ABSTRACT

Corticosteroids are the preeminent antiinflammatory agents although the molecular mechanisms that impart their efficacy have not been defined. The endothelium plays a critical role in inflammation by directing circulating leukocytes into extravascular tissues by expressing adhesive molecules for leukocytes [e.g., endothelial-leukocyte adhesion molecule 1 (ELAM-1) and intercellular adhesion molecule 1 (ICAM-1)]. We therefore determined whether corticosteroids suppress inflammation by inhibiting endothelial expression of adhesion molecules for neutrophils (polymorphonuclear leukocytes). Preincubation of endothelial cells with endotoxin [lipopolysaccharide (LPS), 1 microgram/ml] led to a 4-fold increase in subsequent adherence of polymorphonuclear leukocytes (P < 0.0001, n = 10) to endothelial cells, an increase that was markedly attenuated when endothelial cells were treated with dexamethasone (IC50 < 1 nM, P < 0.0001, n = 6 or 7) during preincubation with LPS. Moreover, the steroid receptor agonist cortisol (10 microM), but not its inactive metabolite tetrahydrocortisol (10 microM), diminished LPS-induced endothelial cell adhesiveness. Further evidence that the action of dexamethasone was mediated through ligation of corticosteroid receptors [human glucocorticoid receptors (hGRs)] was provided by experiments utilizing the steroid antagonist RU-486. RU-486 (10 microM), which prevents translocation of ligated hGR to the nucleus by inhibiting dissociation of hGR from heat shock protein 90, completely aborted the effect of dexamethasone on adhesiveness of endothelial cells (P < 0.0005, n = 3). Treatment of endothelial cells with LPS (1 microgram/ml) stimulated transcription of ELAM-1, as shown by Northern blot analysis, and expression of membrane-associated ELAM-1 and ICAM-1, as shown by quantitative immunofluorescence (both P < 0.001, n = 9). Dexamethasone markedly inhibited LPS-stimulated accumulation of mRNA for ELAM-1 and expression of ELAM-1 and ICAM-1 (IC50 < 10 nM, both P < 0.001, n = 4-9); inhibition of expression by dexamethasone was reversed by RU-486 (both P < 0.005, n = 4-6). As in the adhesion studies, cortisol but not tetrahydrocortisol inhibited expression of ELAM-1 and ICAM-1 (both P < 0.005, n = 3 or 4). In contrast, sodium salicylate (1 mM) inhibited neither adhesion nor expression of these adhesion molecules. These studies suggest that antagonism by dexamethasone of endotoxin-induced inflammation is a specific instance of the general biological principle that the glucocorticoid receptor is a hormone-dependent regulator of transcription.


Subject(s)
Adrenal Cortex Hormones/pharmacology , Adrenal Cortex Hormones/physiology , Anti-Inflammatory Agents/pharmacology , Antigens, CD/biosynthesis , Cell Adhesion Molecules/biosynthesis , Cell Adhesion/physiology , Dexamethasone/pharmacology , Endothelium, Vascular/physiology , Neutrophils/physiology , Receptors, Glucocorticoid/physiology , Antibodies, Monoclonal , Antigens, CD/analysis , Cell Adhesion/drug effects , Cell Adhesion Molecules/analysis , E-Selectin , Endothelium, Vascular/drug effects , Humans , Hydrocortisone/pharmacology , Intercellular Adhesion Molecule-1 , Lipopolysaccharides/pharmacology , Mifepristone/pharmacology , Neutrophils/drug effects , RNA, Messenger/metabolism , Receptors, Glucocorticoid/drug effects , Salmonella typhimurium , Sodium Salicylate/pharmacology , Tetrahydrocortisol/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Umbilical Veins
13.
J Immunol ; 148(7): 2201-6, 1992 Apr 01.
Article in English | MEDLINE | ID: mdl-1347551

ABSTRACT

We have recently demonstrated that human neutrophils (PMN) possess two different classes of adenosine receptors (A1 and A2) that, when occupied, promote chemotaxis and inhibit the generation of reactive oxygen species (e.g., O2- and H2O2), respectively. We have previously demonstrated that adenosine protects endothelial cells (EC) from injury by stimulated neutrophils (PMN) both by diminishing generation of H2O2 and inhibiting adherence of PMN to EC. We therefore determined whether occupancy of A1 or A2 adenosine receptors regulated adherence of PMN to EC. At concentrations similar to those required to inhibit release of O2- by ligation of A2 receptors, both adenosine (IC50 = 56 nM) and 5'N-ethylcarboxamidoadenosine (NECA, IC50 = 8 nM), the most potent A2 agonist, inhibited adherence to EC by stimulated PMN (FMLP, 0.1 microM). In direct contrast, the specific A1 agonists N6-phenylisopropyladenosine and N6-cyclopentyladenosine (CPA) promoted PMN adherence to EC at concentrations of 1-100 nM. To further investigate the mechanisms by which adenosine receptor agonists affected the adherence of stimulated PMN we examined the effect of NECA (A2) and CPA (A1) on the adherence of PMN to fibrinogen (a ligand for the beta 2 integrin CD11b/CD18) and to gelatin. In a dose-dependent manner (IC50 = 2 nM), NECA inhibited the adherence of FMLP-treated PMN to fibrinogen- but not gelatin-coated plates. In contrast, CPA (A1) promoted adherence of stimulated PMN to gelatin-(EC50 = 13 pM) but not fibrinogen-coated plates. Theophylline (10 microM), an adenosine receptor antagonist, reversed the inhibition by NECA (0.3 microM) of stimulated neutrophil adherence to fibrinogen. These observations not only confirm the presence of A1 and A2 receptors on PMN but also suggest two opposing roles for adenosine in inflammation. Occupancy of A1 receptors promotes neutrophil adherence to endothelium and chemotaxis (a proinflammatory role) whereas occupancy of A2 receptors inhibits adherence and generation of toxic oxygen metabolites (an antiinflammatory role).


Subject(s)
Endothelium, Vascular/physiology , Neutrophils/physiology , Receptors, Purinergic/physiology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine-5'-(N-ethylcarboxamide) , Animals , Antigens, CD/physiology , CD18 Antigens , Cell Adhesion , Humans , Macrophage-1 Antigen/physiology , Mice , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Phenylisopropyladenosine/pharmacology , Theophylline/pharmacology
15.
Eur J Orthod ; 13(2): 86-94, 1991 Apr.
Article in English | MEDLINE | ID: mdl-2055257

ABSTRACT

This paper illustrates some of the changes which occur during the treatment of Class II division 1 malocclusions complicated by a deep bite, and reviews the significance of these changes in relation to concepts of deep bite treatment. Particular reference is made to mandibular growth rotation and consequent differential tooth eruption in assessing factors involved in initial bite opening and consolidation of the opened bite. The cases shown illustrate that although an initial bite opening may occur by incisor intrusion and molar eruption, when viewed over a longer period of time rotational mandibular growth and associated differential eruption of teeth in which molars erupt more than incisors may be a more significant factor. Differential eruption which takes place in response to vertical condylar growth under guidance of the appliance would appear to be a significant factor in treatment of deep bite.


Subject(s)
Malocclusion, Angle Class II/therapy , Mandible/growth & development , Vertical Dimension , Activator Appliances , Adolescent , Female , Humans , Male , Mandibular Condyle/growth & development , Orthodontic Appliances , Orthodontic Wires , Rotation , Tooth Eruption, Ectopic/therapy , Tooth Extraction
16.
Proc Natl Acad Sci U S A ; 88(6): 2441-5, 1991 Mar 15.
Article in English | MEDLINE | ID: mdl-2006182

ABSTRACT

Although commonly used to control a variety of inflammatory diseases, the mechanism of action of a low dose of methotrexate remains a mystery. Methotrexate accumulates intracellularly where it may interfere with purine metabolism. Therefore, we determined whether a 48-hr pretreatment with methotrexate affected adenosine release from [14C]adenine-labeled human fibroblasts and umbilical vein endothelial cells. Methotrexate significantly increased adenosine release by fibroblasts from 4 +/- 1% to 31 +/- 6% of total purine released (EC50, 1 nM) and by endothelial cells from 24 +/- 4% to 42 +/- 7%. Methotrexate-enhanced adenosine release from fibroblasts was further increased to 51 +/- 4% (EC50, 6 nM) and from endothelial cells was increased to 58 +/- 5% of total purine released by exposure to stimulated (fMet-Leu-Phe at 0.1 microM) neutrophils. The effect of methotrexate on adenosine release was not due to cytotoxicity since cells treated with maximal concentrations of methotrexate took up [14C]adenine and released 14C-labeled purine (a measure of cell injury) in a manner identical to control cells. Methotrexate treatment of fibroblasts dramatically inhibited adherence to fibroblasts by both unstimulated neutrophils (IC50, 9 nM) and stimulated neutrophils (IC50, 13 nM). Methotrexate treatment inhibited neutrophil adherence by enhancing adenosine release from fibroblasts since digestion of extracellular adenosine by added adenosine deaminase completely abrogated the effect of methotrexate on neutrophil adherence without, itself, affecting adherence. One hypothesis that explains the effect of methotrexate on adenosine release is that, by inhibition of 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) transformylase, methotrexate induces the accumulation of AICAR, the nucleoside precursor of which (5-aminoimidazole-4-carboxamide ribonucleoside referred to hereafter as acadesine) has previously been shown to cause adenosine release from ischemic cardiac tissue. We found that acadesine also promotes adenosine release from and inhibits neutrophil adherence to connective tissue cells. The observation that the antiinflammatory actions of methotrexate are due to the capacity of methotrexate to induce adenosine release may form the basis for the development of an additional class of antiinflammatory drugs.


Subject(s)
Adenosine/metabolism , Connective Tissue/physiology , Endothelium, Vascular/physiology , Methotrexate/pharmacology , Neutrophils/physiology , Adenine/metabolism , Analysis of Variance , Cell Line , Cells, Cultured , Connective Tissue/drug effects , Endothelium, Vascular/drug effects , Fibroblasts/drug effects , Fibroblasts/physiology , Humans , In Vitro Techniques , Kinetics , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Umbilical Veins
18.
Am J Physiol ; 259(4 Pt 2): F598-604, 1990 Oct.
Article in English | MEDLINE | ID: mdl-2221099

ABSTRACT

Male Munich-Wistar rats underwent right nephrectomy and were given weekly injections of deoxycorticosterone acetate (DOCA) and 1% saline (salt) to drink. Two studies were performed. In the first, rats given enalapril (ENP) were compared with controls. In the second, rats ingested either standard chow or chow to which the calcium-entry blocker nifedipine (NIF) had been added. Six to eight weeks after nephrectomy, both control DOCA-salt rats and those given ENP had severe hypertension and significant proteinuria. Rats given NIF excreted less protein, and glomerular lesions were not observed in this group. The effects of NIF on several parameters that have been associated with glomerular injury were examined. Micropuncture studies revealed that glomerular capillary pressure was increased in DOCA-salt rats and was not reduced by NIF. Platelet aggregation was also similar in NIF-treated and control rats. Morphometric studies revealed a tendency toward lower glomerular volume of NIF-treated rats; however, kidney weight and glomerular capillary radius were unaffected by therapy. Thus NIF, but not ENP, prevents DOCA-salt rats from developing hypertension and glomerular injury. This effect does not depend on reduction in glomerular pressure or inhibition of platelet aggregation.


Subject(s)
Desoxycorticosterone , Enalapril/pharmacology , Hypertension/physiopathology , Kidney Glomerulus/physiopathology , Nifedipine/pharmacology , Sodium Chloride , Animals , Hypertension/blood , Hypertension/chemically induced , Hypertension/drug therapy , Hypertension/pathology , Kidney Glomerulus/pathology , Male , Platelet Aggregation , Rats , Rats, Inbred Strains
19.
Arteriosclerosis ; 10(2): 198-207, 1990.
Article in English | MEDLINE | ID: mdl-2317157

ABSTRACT

Patients with chronic renal failure who undergo hemodialysis experience accelerated atherosclerosis and premature death. Since the end-metabolite, oxalic acid, accumulates in plasma in proportion to the severity of renal failure, we studied whether sodium oxalate (0 to 300 microM) is an endothelial toxin and, therefore, might enhance atherogenesis. Exposure to uremic levels of oxalate (greater than 30 microM) for 9 to 28 days depressed endothelial cell replication by 33% to 84% (mean +/- SD, 54% +/- 15.7%, n = 17 experiments, p = 0.002). In contrast, replication of fibroblasts exposed to 200 microM oxalate for 45 days was not inhibited. The inhibitory effect of oxalate on endothelial cell replication was both dose- and time-dependent (both p less than 0.0001) and was first detected 3 to 7 days after the initial exposure to oxalate. Further, the inhibitory effect was fully reversible upon removal of oxalate, but only if exposure was limited to 5 days or less. Sodium salts of other carboxylic acids (citric, succinic, glyoxylic, and malonic; 200 microM) as well as HCl (200 microM) did not suppress endothelial cell replication. Oxalate also inhibited endothelial cell migration but had no effect on basal, thrombin-induced, or arachidonate-induced prostacyclin production by endothelial cells. Exposure of endothelial cells to sodium oxalate (200 microM) for as little as 24 hours-a time period sufficient to induce delayed, transient inhibition of replication not detectable until approximately 1 week after exposure-inhibited incorporation of 3H-leucine into protein by 40% (p = 0.009). We conclude that sodium oxalate acts as a uremic toxin, inhibiting endothelial cell replication and migration, functions which may be important for constitutive inhibition of atherosclerosis.


Subject(s)
Endothelium, Vascular/physiopathology , Oxalates/blood , Uremia/physiopathology , 6-Ketoprostaglandin F1 alpha/biosynthesis , Analysis of Variance , Arteriosclerosis/etiology , Calcium/metabolism , Cell Division , Cell Movement , Cells, Cultured , Chelating Agents , Endothelium, Vascular/drug effects , Endothelium, Vascular/ultrastructure , Humans , Kidney Failure, Chronic/blood , Leucine/metabolism , Oxalates/toxicity , Oxalic Acid , Radioimmunoassay , Thymidine/metabolism , Uremia/blood
20.
Blood ; 74(5): 1635-43, 1989 Oct.
Article in English | MEDLINE | ID: mdl-2529003

ABSTRACT

The observation that aspirin inhibits the increment in tissue plasminogen activator (t-PA) activity induced by venous occlusion of the forearm became controversial with the publication of several nonconfirmatory studies. The current study was performed to confirm the original observation and determine the mechanism by which aspirin suppresses the incremental t-PA activity induced by venous occlusion. Aspirin (650 mg/d X 2) caused no change in resting levels of t-PA antigen (t-PA:Ag) or activity, plasminogen activator inhibitor 1 antigen (PAI-1:Ag), or activity or t-PA-PAI-1 complexes. In contrast, aspirin reduced the increments induced by venous occlusion as follows: t-PA:Ag by 45% (P = .001); t-PA activity (euglobulin lysis time, ELT) by 43% (P = .006); and t-PA activity (alpha 2-plasmin inhibitor-plasmin complexes, PIPC) by 41% (P = .003). The inhibition of incremental t-PA activity measured as ELT or PIPC was linearly correlated with the inhibition of incremental t-PA:Ag (respectively, r = .75, P less than .02; r = .67, P less than .05). Aspirin had no effect on the increment in PAI-1:Ag induced by venous occlusion, but similar to the effect on t-PA:Ag, aspirin induced a 51% inhibition of the increment in t-PA-PAI-1 complex formation. Aspirin did not alter the ability of alpha 2-plasmin inhibitor to bind plasmin, nor the ability of plasma to support the fibrin-catalyzed generation of plasmin by t-PA, nor the subsequent formation of PIPC. Aspirin inhibits the t-PA activity induced by venous occlusion primarily by inhibiting the release of t-PA antigen.


Subject(s)
Antifibrinolytic Agents/metabolism , Aspirin/pharmacology , Fibrinolysin/metabolism , Plasminogen Inactivators/blood , Tissue Plasminogen Activator/antagonists & inhibitors , alpha-2-Antiplasmin , Enzyme-Linked Immunosorbent Assay , Humans , Kinetics , Tissue Plasminogen Activator/analysis , Tissue Plasminogen Activator/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...