Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 141
Filter
1.
J Cardiovasc Pharmacol ; 38(6): 859-67, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11707689

ABSTRACT

Injury during reperfusion can partially offset the benefit of relief of ischemia in myocardial infarctions rapidly treated with thrombolytic drugs or angioplasty. We assessed whether bucillamine (N-[2-mercapto-2-methylpropionyl]-L-cysteine) is potentially useful to treat myocardial reperfusion injury. Bucillamine is a potent sulfhydryl donor not previously tested as a treatment of reperfusion injury. Cardiac myocytes were exposed to hydrogen peroxide or a xanthine/xanthine oxidase system resulting in injury-induced release of lactate dehydrogenase. Bucillamine (125-500 microM) prevented lactate dehydrogenase release in a concentration-dependent manner. Bucillamine, which has two donatable thiol groups, was twice as protective as N-2-mercaptopropionyl glycine, which contains a single donatable thiol group. Dogs were then exposed to 90 min of coronary artery occlusion and 48 h of reperfusion before sacrifice. Beginning at the onset of reperfusion, bucillamine, 11 or 22 mg/kg per hour, or vehicle (saline) was administered intravenously for 3 h. There was a dose-related response to bucillamine for infarct size, normalized for size of the region at risk and adjusted for collateral blood flow to the ischemic region. Infarct size was reduced by 41% in the group treated with bucillamine 22 mg/kg per hour, compared with the vehicle group. Bucillamine, probably through an antioxidant mechanism, reduced infarct size when administered during reperfusion.


Subject(s)
Cysteine/analogs & derivatives , Cysteine/therapeutic use , Myocardial Reperfusion Injury/prevention & control , Animals , Cells, Cultured , Collateral Circulation/drug effects , Coronary Circulation/drug effects , Cysteine/chemistry , Dogs , Dose-Response Relationship, Drug , Hydrogen Peroxide/pharmacology , Kinetics , L-Lactate Dehydrogenase/analysis , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Rats , Single-Blind Method , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/therapeutic use , Tiopronin/chemistry , Tiopronin/therapeutic use
2.
Circulation ; 104(18): 2222-7, 2001 Oct 30.
Article in English | MEDLINE | ID: mdl-11684635

ABSTRACT

Background- Vascular smooth muscle cell (VSMC) proliferation is a critical factor in the neointima formation that causes restenosis after coronary angioplasty (PTCA). Desferri-exochelin 772SM (D-EXO), a highly diffusible, lipophilic iron chelator secreted by Mycobacterium tuberculosis, inhibits proliferation of VSMCs in culture. We hypothesized that treatment with D-EXO would inhibit neointima formation in balloon-injured vessels in vivo. Methods and Results- We subjected 24 pigs to overstretch coronary artery injury with standard PTCA balloons and then administered intramural injections of either D-EXO (n=14) or vehicle (n=10) through an Infiltrator catheter. Treatments were randomized, and the investigators were blinded with regard to treatment group until data analysis was completed. One month later, we euthanized the pigs, excised the injured coronary segments, made multiple sections of each segment, and identified the site of maximal neointima formation. An injury score based on the degree of disruption of the internal or external elastic lamina or media was assigned. D-EXO reduced stenosis index (neointima area divided by the area within the internal elastic lamina), adjusted for injury score, by 47%. Neointima thickness was also reduced. Conclusions- D-EXO, injected intramurally, substantially inhibited formation of neointima in a porcine vascular injury model.


Subject(s)
Angioplasty, Balloon, Coronary , Coronary Restenosis/prevention & control , Iron/metabolism , Peptides, Cyclic/metabolism , Peptides, Cyclic/pharmacology , Angioplasty, Balloon, Coronary/adverse effects , Animals , Coronary Restenosis/etiology , Coronary Restenosis/pathology , Disease Models, Animal , Iron Chelating Agents/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Swine , Treatment Outcome
3.
Transplantation ; 71(1): 112-8, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11211175

ABSTRACT

BACKGROUND: Hepatic ischemia/reperfusion (I/R) injury is associated with early and late graft failure after liver transplantation. A major mechanism is leukocyte adhesion to endothelium followed by release of reactive oxygen intermediates. We examined whether desferriexochelin 772SM (D-Exo), a lipid soluble iron chelator that prevents hydroxyl radical formation, can enhance the capacity of recombinant P-selectin glycoprotein ligand immunoglobulin (rPSGL-Ig), a glycoprotein that binds to P-selectin and inhibits neutrophil adhesion, to protect against I/R injury in an ex vivo rat liver model. METHODS: Rat livers were harvested and stored for 6 hr at 4 degrees C in University of Wisconsin solution and then perfused with oxygenated whole blood for 2 hr. Three groups were studied (n=6 rats/group): an untreated control group; a group that received 0.4 mg/kg rPSGL-Ig intraportally at the time of harvest; and a group that received 0.4 mg/kg rPSGL-Ig plus 1 micromol D-Exo intraportally both at the time of harvest and at the onset of reperfusion. Liver portal venous blood flow was assessed during perfusion, and at the end of each experiment, liver samples were collected for blinded histological evaluation and biochemical analyses. RESULTS: Livers treated with D-Exo + rPSGL-Ig had significantly higher blood flow than livers treated with rPSGL-1Ig alone (P<0.05), and both treatment groups had higher blood flow than controls (P<0.001). Production of carbonyl proteins, a protein oxidation product, was significantly reduced in the D-Exo + rPSGL-1Ig group (P<0.02 vs. controls), but not in the rPSGL-Ig alone group. Total reduced glutathione was significantly higher than controls in the D-Exo + rPSGL-Ig group (P<0.001 vs. controls), but not in the rPSGL-Ig alone group, indicating less oxidative stress in the D-Exo-treated group. Production of malondialdehyde, an index of lipid peroxidation, was significantly less than controls in both treatment groups (P<0.03). Histopathological findings paralleled these results with Banffs scores of 3.3+/-0.5, 1.8+/-0.4, and 1.3+/-0.5 in the control, rPSGL-Ig alone, and D-Exo plus rPSGL-Ig groups, resp. CONCLUSION: rPSGL-Ig provides partial protection against I/R injury to ex vivo rat livers; however, the addition of D-Exo substantially increases protection by reducing oxidative injury. These findings may have clinical relevance in preventing the consequences of I/R injury after liver transplantation.


Subject(s)
Iron Chelating Agents/therapeutic use , Membrane Glycoproteins/therapeutic use , Peptides, Cyclic/therapeutic use , Reperfusion Injury/prevention & control , Animals , Drug Therapy, Combination , Glutathione/analysis , Glutathione Disulfide/analysis , Ligands , Liver/anatomy & histology , Liver/blood supply , Liver/chemistry , Male , Malondialdehyde/analysis , Membrane Glycoproteins/antagonists & inhibitors , Models, Animal , Oxidative Stress/physiology , P-Selectin , Rats , Rats, Sprague-Dawley , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/therapeutic use , Regional Blood Flow/drug effects
4.
Breast Cancer Res Treat ; 69(1): 69-79, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11759830

ABSTRACT

A major goal of cancer chemotherapy is the identification of cytotoxic compounds that are highly selective for cancer cells. We describe here one such compound - a novel iron chelator, desferri-exochelin 772SM. This desferri-exochelin has unique chemical and pharmacological properties, including extremely high iron binding affinity, the capacity to block iron-mediated redox reactions, and lipid solubility which enables it to enter cells rapidly. At low concentrations, this desferri-exochelin kills T47D-YB and MCF-7 human breast cancer cells by inducing apoptosis, but only reversibly arrests the growth of normal human mammary epithelial cells without cytotoxicity. Since iron-loaded exochelin is ineffective, iron chelation accounts for the efficacy of desferri-exochelin. For both the killing of breast cancer cells and the growth arrest of normal breast epithelial cells, desferri-exochelin was effective at much lower concentrations than the lipid-insoluble iron chelator deferoxamine, which has shown only limited potential as an anti-cancer agent. Growth arrest of progesterone receptor positive T47D-YB cells with the progestin R5020 transiently protects them from the cytotoxic effects of desferri-exochelin, but the cells are killed after cell growth resumes. Similarly, MCF-7 cells arrested with the estrogen antagonist ICI182780 are transiently resistant to killing by desferri-exochelin. Thus the desferri-exochelin is cytotoxic only to actively growing tumor cells. Since desferri-exochelin 772SM can selectively and efficiently destroy proliferating cancer cells without damaging normal cells, it may prove useful for the treatment of cancer.


Subject(s)
Apoptosis , Breast Neoplasms/pathology , Breast/cytology , Iron Chelating Agents/pharmacology , Peptides, Cyclic/pharmacology , Cell Division , Dose-Response Relationship, Drug , Female , Humans , Tumor Cells, Cultured
5.
J Biol Chem ; 275(23): 17821-6, 2000 Jun 09.
Article in English | MEDLINE | ID: mdl-10748174

ABSTRACT

Proliferation of vascular smooth muscle cells (VSMC) is characteristic of restenosis following balloon angioplasty. We show here that a low concentration of a novel iron chelator, desferri-exochelin 772SM, reversibly arrests the growth of human VSMC in vitro, specifically in G(0)/G(1) and S phases. The lipophilic desferri-exochelin is effective more rapidly and at a 10-fold lower concentration than the nonlipophilic iron chelator deferoxamine. Treatment of growth-synchronized VSMC with the desferri-exochelin results in down-regulation of cyclin E/ Cdk2 and cyclin A/Cdk2 activity but does not affect the cyclin D/Cdk4/retinoblastoma phosphorylation pathway. Both DNA replication and RNA transcription are inhibited in exochelin-treated cells, but protein synthesis is not. The ability of desferri-exochelin 772SM to reversibly block the growth of VSMC in vitro with no apparent cytotoxicity suggests that the exochelin may be useful as a therapeutic agent to limit restenosis in injured vessels.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle/drug effects , Ferric Compounds/pharmacology , Iron Chelating Agents/pharmacology , Muscle, Smooth, Vascular/cytology , Mycobacterium tuberculosis , Peptides, Cyclic/pharmacology , Aorta , Cell Division/drug effects , Cells, Cultured , Cyclin E/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/metabolism , Deferoxamine/pharmacology , Dose-Response Relationship, Drug , Epidermal Growth Factor/pharmacology , Humans , Iliac Artery , Kinetics , Muscle, Smooth, Vascular/drug effects , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Retinoblastoma Protein/metabolism , Saphenous Vein
6.
Circulation ; 101(15): 1792-8, 2000 Apr 18.
Article in English | MEDLINE | ID: mdl-10769279

ABSTRACT

BACKGROUND: Estrogens have vascular effects through the activation of estrogen receptors (ERs). In addition to ERalpha, the first ER to be cloned, a second subtype called ERbeta has recently been discovered. METHODS AND RESULTS: Using a reverse-transcriptase polymerase chain reaction assay that employs the same primer pair to simultaneously amplify ERalpha and ERbeta transcripts, we found that ERbeta is the ER form that is predominantly expressed in human vascular smooth muscle, particularly in women. The transcriptional effects of the 2 ERs in transfected HeLa cells differed. In response to 17beta-estradiol, ERalpha is a stronger transactivator than ERbeta at low receptor concentrations. However, at higher receptor concentrations, ERalpha activity self-squelches, and ERbeta is a stronger transactivator. Tamoxifen has partial agonist effects with ERalpha but not with ERbeta. CONCLUSIONS: The protective effects of estrogens in the cardiovascular system of women may be due to the genomic effects of ERbeta in vascular tissue.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Transcription, Genetic , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Fulvestrant , HeLa Cells , Humans , Ligands , Promoter Regions, Genetic , RNA/analysis , Receptors, Estrogen/drug effects , Regression Analysis , Tamoxifen/pharmacology
7.
Vasc Med ; 4(2): 93-9, 1999.
Article in English | MEDLINE | ID: mdl-10406456

ABSTRACT

Iron is an essential element for normal cellular function and general health. However, iron may play a pathologic role in certain cardiac conditions including reperfusion injury, hemochromatosis, beta-thalassemia and coronary atherosclerosis. It also may play a role in injury due to anthracycline cardiotoxicity. Removal of iron via phlebotomy for hemochromatosis and chelation therapy for beta-thalassemia are proven treatments. Cell culture, and isolated organ and animal studies suggest that depleting iron stores may prevent reperfusion injury, restenosis and even atherogenesis. This article will review mechanisms by which iron overload states and normal iron stores contribute to cardiovascular pathophysiology and the accumulating evidence that iron chelation may prevent restenosis and atherogenesis.


Subject(s)
Arteriosclerosis/metabolism , Heart Diseases/metabolism , Iron Overload/metabolism , Myocardial Reperfusion Injury/metabolism , Hemochromatosis/metabolism , Humans , beta-Thalassemia/metabolism
8.
J Steroid Biochem Mol Biol ; 69(1-6): 45-50, 1999.
Article in English | MEDLINE | ID: mdl-10418980

ABSTRACT

Pharmacological antagonists of steroid receptor action had been thought to exert their effects by a passive mechanism driven principally by the ability of the antagonist to compete with agonist for the ligand binding site. However, recent analyses of antagonist-occupied receptor function suggest a more complex picture. Antagonists can be subdivided into two groups, type I, or pure antagonists, and type II, or mixed antagonists that can have variable transcriptional activity based upon differential dimerization and DNA binding properties. This led us to propose that receptor antagonism may not simply be a passive competition for the ligand binding site, but may, in some cases, involve active recruitment of corepressor or coactivator proteins to produce a mixed transcriptional phenotype. We used a yeast two-hybrid screen to identify proteins that interact specifically with antagonist-occupied receptors. Two proteins have been characterized: L7/SPA, a ribosome-associated protein that is localized in both the cytoplasm and nucleus, but with no known extranucleolar nuclear function; and hN-CoR, the human homolog of the mouse thyroid receptor corepressor mN-CoR. In in vivo transcription assays we show that L7/SPA enhances the partial agonist activity of type II mixed antagonists, and that N-CoR and the related corepressor, SMRT, suppresses it. The coregulators do not affect agonists or pure antagonists. Moreover, the net agonist activity seen with mixed antagonists is a function of the ratio of coactivator to corepressor. Based upon these results, we proposed that in breast tumors the inappropriate agonist activity seen with therapeutic antagonists such as tamoxifen is responsible for the hormone-resistant state. To confirm this, we are quantitating coactivator/corepressor ratios in breast tumor cells lines and clinical breast cancers. Results should provide new insights into the mechanisms underlying the progression of breast cancer to hormone resistance, and may suggest strategies for delaying or reversing this process.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/physiopathology , Receptors, Steroid/antagonists & inhibitors , Tamoxifen/pharmacology , Transcription, Genetic , Animals , Drug Resistance, Neoplasm , Humans , Receptors, Steroid/genetics
9.
Circulation ; 99(20): 2688-93, 1999 May 25.
Article in English | MEDLINE | ID: mdl-10338464

ABSTRACT

BACKGROUND: Estrogens stimulate growth of breast or uterine cells but have the opposite effect on vascular smooth muscle cells, in which they protect against coronary artery disease with or without concomitant administration of progesterone. A possible cause of differences in hormone action is variable tissue-specific expression of hormone receptor. Therefore, we analyzed the structure of estrogen receptors (ERs) and progesterone receptors (PRs) in human vascular smooth muscle. METHODS AND RESULTS: RNA was isolated from human vascular smooth muscle, and the functional domains of ER-alpha and PR were characterized by reverse transcriptase and polymerase chain reaction. Interestingly, in addition to wild-type ER-alpha and PR, 5 variant ER-alpha and 2 variant PR transcripts were found. These variants contained precise deletions of exons encoding regions of the hormone-binding domain. The PR transcripts lacked exon 4 (PRDelta4) and exon 6 (PRDelta6). The ER-alpha transcripts were missing exon 4 (ERDelta4), exon 5 (ERDelta5), exon 6 (ERDelta6), exon 7 (ERDelta7), and exons 6 and 7, (ERDelta6,7). ER-beta variants were also detected. The PR variants were functionally characterized, and PRDelta6 was found to be a dominant-negative transcription inhibitor of wild-type receptors. Variant PR was present in premenopausal women but absent in postmenopausal women. CONCLUSIONS: Variant PR and ER transcripts are extensively expressed in human vascular smooth muscle. The complex tissue-specific effects of sex hormones may be mediated by the expression of heterogeneous forms of their cognate receptors. The presence of variant ERs and PRs may be of importance in altering the physiological effects of estrogens or progestins in vascular smooth muscle.


Subject(s)
Genetic Variation , Muscle, Smooth, Vascular/metabolism , RNA, Messenger/metabolism , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Adult , Aged , Culture Techniques , Estrogen Receptor beta , Exons/genetics , Female , Gene Deletion , Genetic Variation/physiology , Humans , Male , Middle Aged , Postmenopause/metabolism , Premenopause/metabolism , Reverse Transcriptase Polymerase Chain Reaction
10.
J Cardiovasc Pharmacol ; 33(1): 19-29, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9890392

ABSTRACT

Early reperfusion of acute myocardial infarctions halts cell death due to ischemia but causes further injury, probably by oxidant mechanisms. We identified the window of opportunity during which antioxidants must be present in therapeutic concentrations to prevent reperfusion injury during 90 min of ischemia and 48 h of reperfusion in 57 dogs. We examined the effect on myocardial infarct size of intravenous infusion of N-2-mercaptopropionyl glycine (MPG), a diffusible antioxidant with a plasma half-time of 7 min, by using a series of protocols with a range of timing. Whereas infusions of MPG for > or =3 h reduced infarct size by approximately 50%, infusions for 1 h only (the first, second or third hours of reperfusion) caused only small reductions. A statistical analysis that focused on identifying components of group membership responsible for differences revealed that duration of treatment was a major determinant of infarct size. If begun any time within the first hour of reperfusion, infusions of > or =3 h markedly diminished infarct size. Because reperfusion injury proceeds for the first 3 h of reperfusion, but decreases thereafter, adequate protection is needed for > or =3 of the first 4 h of reperfusion, but more prolonged protection is not necessary.


Subject(s)
Myocardial Reperfusion Injury/prevention & control , Tiopronin/therapeutic use , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Dogs , Female , Hemodynamics/drug effects , Male , Myocardial Infarction/complications , Myocardial Reperfusion Injury/etiology , Outcome Assessment, Health Care , Time Factors , Tiopronin/pharmacology
11.
Circulation ; 98(19): 2049-54, 1998 Nov 10.
Article in English | MEDLINE | ID: mdl-9808604

ABSTRACT

BACKGROUND: Estrogen-mediated cardiovascular protection is incompletely explained by its beneficial lipid-modifying effects. Previous studies interrogating direct vascular effects of estrogens have used models of either diet- or injury-induced atherosclerosis. The purpose of this study was to determine the influence of ovarian ablation alone on vascular remodeling. We hypothesized that estrogens are atheroprotective, independent of their influence on lipid metabolism, by directly influencing the production and effects of a prototypical atherogenic mediator, basic fibroblast growth factor (bFGF). METHODS AND RESULTS: Twenty-five female sheep were randomized to sham operation, ovariectomy, or ovariectomy plus 17beta-estradiol replacement. Serum cholesterol and triglyceride levels were serially measured for 1 year and were similar among groups and in the normal range (30 to 60 mg/dL). At 6, 9, and 12 months, ovariectomy resulted in aortoiliac intimal hyperplasia compared with sham (P<0.01) and hormone replacement (P<0.01) groups. The neointima of ovariectomized animals was characterized immunohistochemically by increased vascular smooth muscle cells (VSMCs). Levels of bFGF protein were determined in adjacent aortic segments. Ovariectomized sheep had 2-fold more FGF than sham or ovariectomized sheep that received hormone replacement. In vitro, estradiol inhibited the mitogenic effect of bFGF on human aortic VSMCs. CONCLUSIONS: Without dietary manipulation, ovarian ablation alone induces aortic intimal hyperplasia in the ewe. Estradiol abrogates this response independently of its effects on serum lipids. Hormone replacement decreases the accumulation of the atherogenic peptide bFGF in vivo and inhibits the mitogenic response of VSMCs to bFGF in vitro. These results suggest that estrogens may provide atheroprotection both by modulating local production and by attenuating the influence of bFGF on VSMC growth.


Subject(s)
Aorta/metabolism , Aorta/pathology , Fibroblast Growth Factors/metabolism , Ovariectomy/adverse effects , Tunica Intima/pathology , Animals , Aorta/drug effects , Cell Division/drug effects , Estradiol/pharmacology , Female , Fibroblast Growth Factors/antagonists & inhibitors , Humans , Hyperplasia , Immunohistochemistry , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Sheep
14.
Proc Natl Acad Sci U S A ; 95(9): 5263-8, 1998 Apr 28.
Article in English | MEDLINE | ID: mdl-9560264

ABSTRACT

Reperfusion injury, which occurs upon the reintroduction of blood flow to an ischemic organ, is responsible for considerable damage in heart attacks and strokes. However, no treatment for reperfusion injury is currently available. A major cause of reperfusion injury is the iron-mediated generation of hydroxyl radical (.OH). In this study we have explored the capacity of novel iron chelators called "exochelins" to prevent reperfusion injury. Exochelins, siderophores of Mycobacterium tuberculosis, are unique iron chelators because they are lipid soluble, and hence able to enter cells rapidly. In the iron-free state, exochelins prevented .OH formation. Desferri-exochelins prevented oxidative injury to cultured cardiac myocytes, and did so more rapidly and effectively than the nonlipid soluble iron chelator deferoxamine. The capacity of various desferri-exochelins to protect myocytes from oxidative injury varied directly with their solubility in lipid. Infused into isolated rabbit hearts during reperfusion after a period of ischemia, desferri-exochelins dramatically improved systolic and diastolic left ventricular function, preserved coronary flow, reduced release of the cardiac enzyme lactic dehydrogenase, and reduced myocardial concentrations of .OH metabolites. Thus, highly diffusible desferri-exochelins block injury caused by .OH production and have potential for the treatment of reperfusion injury.


Subject(s)
Myocardial Reperfusion Injury/prevention & control , Peptides, Cyclic/pharmacology , Siderophores/pharmacology , Animals , Cells, Cultured , Hydrogen Peroxide/toxicity , Male , Mycobacterium tuberculosis/chemistry , Peptides, Cyclic/chemistry , Rabbits , Rats , Reactive Oxygen Species , Siderophores/chemistry , Solubility , Superoxides/toxicity
16.
J Surg Res ; 80(2): 380-5, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9878341

ABSTRACT

BACKGROUND: The role of estrogens in providing atheroprotection has been well documented in both epidemiologic and experimental studies. This phenomenon has traditionally been attributed to the beneficial lipid-modifying effects of estrogens. Previous studies have used models of either diet- or injury-induced atherosclerosis. As such, the interrelationship between estrogens, lipids, and atherosclerosis remains unclear. We hypothesized that estrogens are atheroprotective independent of changes in serum lipids by directly influencing the accumulation and effects of the peptide growth factor transforming growth factor beta1 (TGF-beta1). MATERIAL AND METHODS: Thirteen female sheep (8 years old) were randomized to sham, ovariectomy, or ovariectomy with 17beta-estradiol replacement. Serum lipid levels were serially measured. At 9 months, necropsy was performed with histologic morphometric analysis of the aortoiliac bifurcation. Levels of TGF-beta1 were determined in serum and aortic tissue. Human aortic smooth muscle cells were isolated and cultured. RESULTS: Serum triglyceride, lipoprotein a, and total, low-density lipoprotein, and high-density lipoprotein cholesterol levels were similar and normal between groups. Ovariectomy resulted in aortoiliac intimal hyperplasia compared with sham (P < 0.001) and hormone replacement (P < 0.001) groups. Compared with ovariectomy, estrogen replacement attenuated aortic accumulation of TGF-beta1 (P < 0.02). In vitro, estradiol potentiated TGF-beta1 inhibition of human vascular smooth muscle cell (VSMC) proliferation and increased TGF-beta1 release in stimulated VSMCs (P < 0.001). CONCLUSIONS: Without dietary manipulation, ovarian ablation induces aortic intimal hyperplasia in the ewe. Estradiol abrogates this response independently of its effects on serum lipids. Hormone replacement decreases the accumulation of TGF-beta1, suggesting that estrogens may provide atheroprotection both by modifying local production and by modulating the influence of TGF-beta1 on VSMC growth.


Subject(s)
Arteriosclerosis/prevention & control , Estrogen Replacement Therapy , Transforming Growth Factor beta/metabolism , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Arteriosclerosis/metabolism , Arteriosclerosis/pathology , Cell Division/drug effects , Estradiol/pharmacology , Female , Humans , Hyperplasia , Lipids/blood , Luteinizing Hormone/blood , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Ovariectomy , Sheep , Transforming Growth Factor beta/blood
17.
Am J Hypertens ; 10(11): 1263-9, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9397246

ABSTRACT

The comparative effects of the once a day calcium channel antagonists amlodipine and long-acting diltiazem were assessed in a parallel design, investigator-blinded, multicenter trial in 123 patients with diastolic blood pressures ranging from 95 to 114 mm Hg before treatment. Patients were randomized to one of the two drugs and titrated at 2-week intervals to 5 or 10 mg of amlodipine or 180, 240, or 360 mg of long-acting diltiazem during a 10-week treatment period. Both drugs significantly reduced resting, sitting, standing, and 24-h ambulatory systolic and diastolic pressures. Amlodipine caused significantly greater reductions in sitting and standing systolic pressures, standing diastolic pressures, and 24-h ambulatory systolic and diastolic pressures versus diltiazem. Sitting systolic pressures were reduced from 151.9 +/- 2.0 (SE) at baseline to 137.9 +/- 1.8 mm Hg with amlodipine treatment and from 149.0 +/- 2.1 to 145.1 +/- 2.5 mm Hg with diltiazem. Sitting diastolic pressures were reduced from 100.2 +/- 0.6 to 87.8 +/- 1.0 mm Hg with amlodipine and from 101.1 +/- 1.0 to 91.9 +/- 1.1 mm Hg with diltiazem. Reductions in standing systolic pressures after treatment were -12.1 +/- 1.5 mm Hg amlodipine v -4.6 +/- 1.5 mm Hg diltiazem (P < .01), and reductions in standing diastolic pressures were -11.8 +/- 0.9 mm Hg amlodipine v -8.6 +/- 0.9 mm Hg diltiazem (P < .02). Heart rates did not change significantly with either drug during the study. Two subjects in each group dropped out because of adverse experiences. Although both agents were well tolerated and reduced blood pressures consistently over the 10-week test period, amlodipine was more effective than diltiazem in reducing systolic and diastolic blood pressures to the target pressures of < 140 mm Hg systolic and < 90 mm Hg diastolic over a range of doses widely used in clinical practice.


Subject(s)
Amlodipine/therapeutic use , Antihypertensive Agents/therapeutic use , Diltiazem/therapeutic use , Hypertension/drug therapy , Adult , Aged , Aged, 80 and over , Amlodipine/adverse effects , Antihypertensive Agents/adverse effects , Blood Pressure/drug effects , Blood Pressure Monitoring, Ambulatory , Diltiazem/adverse effects , Double-Blind Method , Electrocardiography , Female , Heart Rate/drug effects , Humans , Hypertension/physiopathology , Male , Middle Aged , Patient Compliance , Patient Dropouts
18.
Am J Physiol ; 272(2 Pt 2): H618-24, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9124417

ABSTRACT

Ischemia and reperfusion cause coronary vascular and myocardial injury, which may be due to leukocyte-mediated processes. Antileukocyte measures have reduced injury after brief reperfusion periods of 1-3 h, but there has been little information on whether benefits are apparent after longer periods of reperfusion. We examined the effect of pretreatment with a monoclonal antibody (R15.7) to the CD18 family of leukocyte adhesion molecules (beta2-integrins) in dogs exposed to regional coronary ischemia for 1 h of left anterior descending coronary artery ligation and then reperfused for 48 h. Coronary microvascular permeability was assessed in vivo by measurement of protein leak index (PLI), using a double-isotope technique with autologous radiolabeled transferrin and erythrocytes. Vasorelaxation was measured in vitro with preconstricted epicardial coronary artery rings subjected to increasing concentrations of the endothelium-dependent vasodilators bradykinin (BK) and ADP and the endothelium-independent vasodilator nitroprusside. At 48 h of reperfusion in untreated dogs there were substantial increases in PLI in the previously ischemic regions, indicative of increased extravascular transferrin. These abnormalities were decreased, but not abolished, in the dogs treated with R15.7. Relaxation of rings from the ischemic/reperfused artery to BK and ADP were blunted in the untreated dogs. R15.7 resulted in improvement in some, but not all, indexes of relaxation in response to BK and ADP. Relaxation to nitroprusside was normal in ischemic/reperfused coronary rings from both treated and untreated dogs. Therefore, after 1 h of regional coronary ischemia and 48 h of reperfusion, coronary endothelial injury, which was manifested by increased coronary microvascular permeability and abnormalities in coronary endothelium-dependent relaxation, was reduced by pretreatment with the anti-CD18 integrin antibody R15.7.


Subject(s)
Antibodies/pharmacology , Coronary Vessels/pathology , Integrins/immunology , Leukocytes/metabolism , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/pathology , Animals , Antibodies/immunology , Blood Proteins/metabolism , Capillary Permeability , Coronary Vessels/drug effects , Coronary Vessels/physiopathology , Dogs , In Vitro Techniques , Male , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/physiopathology , Neutrophils/immunology , Pericardium/physiopathology , Vasodilation
19.
Circulation ; 94(3): 542-6, 1996 Aug 01.
Article in English | MEDLINE | ID: mdl-8759100

ABSTRACT

BACKGROUND: Polymorphonuclear leukocytes, particularly neutrophils, are important mediators of ischemia/reperfusion-induced myocardial and coronary vascular injury. The selectin family of glycoprotein receptors mediates neutrophil "rolling," a loose, transient adhesion to the coronary endothelium that precedes the firmer adhesion associated with cardiovascular injury. The oligosaccharide sialyl-Lewis(x) (SLe(x)) is the probable neutrophil counterligand for endothelial E- and P-selectin. Administration of analogues of SLe(x) could potentially prevent neutrophil rolling by competing for the selectin-adhesion sites. We investigated the effects of treatment with an analogue of SLe(x) in a chronic canine model of ischemia/reperfusion. METHODS AND RESULTS: Anesthetized mongrel dogs were subjected to 90 minutes of ischemia through occlusion of the left anterior descending coronary artery and 48 hours of reperfusion. Five minutes before the onset of reperfusion, dogs received either the SLe(x) analogue CY-1503 at a dose of 20 mg/kg or normal saline. Myocardial infarct size was measured through triphenyltetrazolium chloride staining, and polymorphonuclear leukocyte accumulation was evaluated through measurement of cardiac myeloperoxidase activity. After adjustment for blood flow, the mean infarct size of control dogs (44.7 +/- 4.2%) was not different from that of treated dogs (33.4 +/- 4.0%, P = .06), although there was a trend toward a slightly lower value in the treated dogs. Myeloperoxidase activity was not different in the infarcted myocardium of the treated group compared with that of the control group (2.7 +/- 0.71 treated versus 1.08 +/- 0.41 units/mg protein control, P = .06). CONCLUSIONS: We conclude that CY-1503 does not substantially or consistently reduce myocardial infarct size or neutrophil accumulation in dogs subjected to ischemia followed by a prolonged period (48 hours) of reperfusion.


Subject(s)
Myocardial Infarction/etiology , Myocardial Infarction/pathology , Myocardial Ischemia/complications , Myocardial Reperfusion , Oligosaccharides/pharmacology , Animals , Coronary Circulation , Dogs , Myocardial Infarction/physiopathology , Myocardial Ischemia/physiopathology , Myocardium/enzymology , Peroxidase/metabolism
20.
Free Radic Biol Med ; 21(6): 743-53, 1996.
Article in English | MEDLINE | ID: mdl-8902520

ABSTRACT

We examined the efficacy of a group of drugs that stabilize the cell membrane and can potentially prevent cytotoxicity in cultured fetal chick cardiac myocytes exposed to hydrogen peroxide (H2O2). The effects of various membrane-protective agents were determined by analysis of the kinetics of lactic dehydrogenase (LDH) release. The kinetic parameters calculated from the data include a rate constant for release of LDH (kb) and the fraction of total LDH that is released from the cells (CIIMax). The CIIMaxs derived from a range of H2O2 concentrations reveal that the mean toxic concentration of H2O2 is 1.1 mM and that the pattern of toxicity is consistent with the damage being directly proportional to the concentration of the free radicals generated from the H2O2. Maximum nontoxic concentrations of three amphiphilic membrane protective agents had no effect upon cytotoxicity from H2O2. The slightly polar lipophilic agent, Trolox C, a vitamin E derivative, was also without protective effect at a maximum nontoxic concentration. The highly lipophilic agent, probucol, had a small protective effect at 50 microM, the maximum concentration we succeeded in solubilizing in the culture medium. However, the lipophilic 21-aminosteroid U74500, delivered to the cells in an emulsion, markedly reduced cytotoxicity from H2O2. The CII Max was significantly reduced and the protection was concentration dependent over a range of concentrations from 50-400 nmol/ml. Furthermore, the inhibition by U74500 was fully consistent with a mechanism of scavenging of free radicals formed during lipid peroxidation. In support of this hypothesis, a dose of 400 nmoles/ml completely prevented an increase in lipid peroxides due to H2O2 exposure, whereas there was a sixfold increase during exposure to H2O2 in untreated myocytes. Thus, a lipid soluble 21-aminosteroid prevented lipid peroxidation and reduced cardiac myocyte injury during exposure to H2O2, probably by scavenging of free radicals formed during lipid peroxidation in the cell membrane, whereas amphiphilic agents, which probably altered the physicochemical structure of the cell membrane but did not scavenge free radicals, were not protective.


Subject(s)
Antioxidants/pharmacology , Cell Membrane/drug effects , Hydrogen Peroxide/toxicity , Lipids , Pregnatrienes/pharmacology , Solubility , Animals , Cell Death/drug effects , Cells, Cultured , Chick Embryo , Chromans/pharmacology , Diltiazem/pharmacology , Dose-Response Relationship, Drug , Free Radical Scavengers , Kinetics , L-Lactate Dehydrogenase/metabolism , Lidocaine/pharmacology , Lipid Peroxidation/drug effects , Probucol/pharmacology , Propranolol/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL